WO2020130077A1 - Composition for removing pluripotent stem cells and method of removing pluripotent stem cells - Google Patents

Composition for removing pluripotent stem cells and method of removing pluripotent stem cells Download PDF

Info

Publication number
WO2020130077A1
WO2020130077A1 PCT/JP2019/049837 JP2019049837W WO2020130077A1 WO 2020130077 A1 WO2020130077 A1 WO 2020130077A1 JP 2019049837 W JP2019049837 W JP 2019049837W WO 2020130077 A1 WO2020130077 A1 WO 2020130077A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
stem cells
pluripotent stem
brq
mouse
Prior art date
Application number
PCT/JP2019/049837
Other languages
French (fr)
Japanese (ja)
Inventor
亨 近藤
Original Assignee
国立大学法人北海道大学
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 国立大学法人北海道大学 filed Critical 国立大学法人北海道大学
Priority to US17/414,511 priority Critical patent/US20220364065A1/en
Priority to EP19898801.6A priority patent/EP3901252A4/en
Priority to JP2020561509A priority patent/JP7409670B2/en
Publication of WO2020130077A1 publication Critical patent/WO2020130077A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/0004Oxidoreductases (1.)
    • C12N9/001Oxidoreductases (1.) acting on the CH-CH group of donors (1.3)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y103/00Oxidoreductases acting on the CH-CH group of donors (1.3)
    • C12Y103/05Oxidoreductases acting on the CH-CH group of donors (1.3) with a quinone or related compound as acceptor (1.3.5)
    • C12Y103/05002Dihydroorotate dehydrogenase (1.3.5.2)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/0081Purging biological preparations of unwanted cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0603Embryonic cells ; Embryoid bodies
    • C12N5/0606Pluripotent embryonic cells, e.g. embryonic stem cells [ES]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0618Cells of the nervous system
    • C12N5/0623Stem cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0696Artificially induced pluripotent stem cells, e.g. iPS
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/99Enzyme inactivation by chemical treatment
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y103/00Oxidoreductases acting on the CH-CH group of donors (1.3)
    • C12Y103/01Oxidoreductases acting on the CH-CH group of donors (1.3) with NAD+ or NADP+ as acceptor (1.3.1)
    • C12Y103/01014Dihydroorotate dehydrogenase (NAD+) (1.3.1.14)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y103/00Oxidoreductases acting on the CH-CH group of donors (1.3)
    • C12Y103/01Oxidoreductases acting on the CH-CH group of donors (1.3) with NAD+ or NADP+ as acceptor (1.3.1)
    • C12Y103/01015Dihydroorotate dehydrogenase (NADP+) (1.3.1.15)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y103/00Oxidoreductases acting on the CH-CH group of donors (1.3)
    • C12Y103/98Oxidoreductases acting on the CH-CH group of donors (1.3) with other, known, acceptors (1.3.98)
    • C12Y103/98001Dihydroorotate oxidase (fumarate) (1.3.98.1)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/32Amino acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/44Thiols, e.g. mercaptoethanol
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/11Epidermal growth factor [EGF]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/115Basic fibroblast growth factor (bFGF, FGF-2)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/235Leukemia inhibitory factor [LIF]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/70Enzymes
    • C12N2501/73Hydrolases (EC 3.)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/02Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from embryonic cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/45Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from artificially induced pluripotent stem cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer

Definitions

  • the present invention relates to a composition for removing pluripotent stem cells and a method for removing pluripotent stem cells. More specifically, the present invention relates to a composition and method for removing undifferentiated pluripotent stem cells remaining in a cell group in which differentiation is induced from pluripotent stem cells.
  • Pluripotent stem cells such as embryonic stem cells (ES cells) and induced pluripotent stem cells (iPS cells) have the ability to differentiate into all the cells that make up the living body, and therefore they are used as the key to the realization of regenerative medicine. Is expected. In addition, until now, various methods for inducing differentiation from these pluripotent stem cells to specific functional cells for transplantation required for treatment have been established.
  • ES cells embryonic stem cells
  • iPS cells induced pluripotent stem cells
  • Non-Patent Documents 1 to 5 various methods for removing pluripotent stem cells have been developed to solve this problem.
  • the present invention has been made in view of the above problems of the prior art, and finds a compound that brings cytotoxicity to undifferentiated pluripotent stem cells, while having little toxicity to other cells.
  • the purpose is to Furthermore, it is an object of the present invention to provide a composition for removing pluripotent stem cells containing the compound as an active ingredient, and a method for removing pluripotent stem cells using the compound.
  • DHODH dihydroorotate dehydrogenase
  • pluripotent stem cells by treating pluripotent stem cells with a DHODH inhibitor and then transplanting them into a mouse, or by administering a DHODH inhibitor to a mouse into which a pluripotent stem cells are transplanted, a particular side effect is brought about in the mouse. It was also confirmed that the tumor formation from pluripotent stem cells could be suppressed without the above.
  • DHODH is an oxidoreductase that catalyzes the fourth chemical reaction of de novo synthesis of pyrimidine. It has already been clarified that the DHODH inhibitor suppresses the proliferation of T cells and B cells and exerts an immunosuppressive effect by inhibiting the synthesis thereof, and the drug is due to such an effect, rheumatoid arthritis and the like. Is used for the treatment of autoimmune diseases of. Furthermore, DHODH inhibitors exert a therapeutic effect on leukemia (acute myeloid leukemia, chronic myelogenous leukemia) by releasing differentiation inhibition in cancer stem cells or by enhancing p53 synthesis in the same cells. It has also been reported (Non-patent documents 6 and 7).
  • the present inventor revealed for the first time that the drug can remove pluripotent stem cells, as described above. ..
  • somatic stem cells neural stem cells, etc.
  • somatic stem cells which are inferior to pluripotent stem cells but have pluripotency and self-proliferative ability and are also classified as stem cells
  • a DHODH inhibitor is The present inventors have found that they do not show significant cytotoxicity, and have completed the present invention.
  • the present invention relates to a composition for removing pluripotent stem cells and a method for removing pluripotent stem cells using a DHODH inhibitor, and more specifically provides the following.
  • the pluripotent stem cell is at least one pluripotent stem cell selected from the group consisting of embryonic stem cells (ES cells), induced pluripotent stem cells (iPS cells) and embryonic tumor cells (EC cells)
  • ES cells embryonic stem cells
  • iPS cells induced pluripotent stem cells
  • EC cells embryonic tumor cells
  • a method for removing residual undifferentiated pluripotent stem cells from the cell group which comprises a step of contacting a cell group differentiated from the pluripotent stem cells with a dihydroorotate dehydrogenase inhibitor.
  • the pluripotent stem cell is at least one pluripotent stem cell selected from the group consisting of embryonic stem cells (ES cells), induced pluripotent stem cells (iPS cells) and embryonic tumor cells (EC cells)
  • ES cells embryonic stem cells
  • iPS cells induced pluripotent stem cells
  • EC cells embryonic tumor cells
  • ⁇ 7> The method according to ⁇ 5> or ⁇ 6>, wherein the dihydroorotic acid dehydrogenase inhibitor is brequinar.
  • the cell group is a cell group containing somatic stem cells differentiated from the pluripotent stem cells.
  • undifferentiated pluripotent stem cells can be removed without causing significant cytotoxicity to differentiated cells. That is, according to the present invention, it becomes possible to remove the undifferentiated pluripotent stem cells remaining in the cell group differentiated from the pluripotent stem cells.
  • FIG. 1 is a graph showing the survival rate of mouse embryonic stem (ES) cells after culturing for 3 days in the presence of a dihydroorotic acid dehydrogenase (DHODH) inhibitor.
  • DHODH dihydroorotic acid dehydrogenase
  • 3 is a fluorescence micrograph showing the results of mixed culture of mouse neural stem cells and mouse ES cells for 3 days in the presence of BRQ.
  • control indicates the culture result in the absence of BRQ.
  • the upper three photographs show the results of detection of a neural stem cell marker (Nestin) by immunostaining.
  • the middle three photographs show the results of detecting pluripotent stem cell markers (Nanog) by immunostaining.
  • the bottom three photographs are photographs in which the results of detecting Nestin and Nanog by immunostaining and the results of counterstaining the cell nuclei with DAPI are superimposed.
  • FIG. 3 is a fluorescence micrograph showing the results of mixed culture of mouse neural stem cells and mouse iPS cells for 3 days in the presence of BRQ.
  • control indicates the culture result in the absence of BRQ.
  • the upper three photographs show the results of detecting a pluripotent stem cell marker (Nanog) by immunostaining.
  • the middle three photographs show the results of detecting a neural stem cell marker (Nestin) by immunostaining.
  • the bottom three photographs are photographs in which the results of detecting Nestin and Nanog by immunostaining and the results of counterstaining the cell nuclei with DAPI are superimposed.
  • FIG. 9 is a graph showing the survival rate of mouse iPS cells when various ribonucleosides (uridine, adenosine, guanosine or cytidine) were added in the presence of 10 ⁇ M BRQ.
  • 6 is a graph showing the survival rate of mouse ES cells when various ribonucleosides (uridine, adenosine, guanosine or cytidine) were added in the presence of 10 ⁇ M BRQ.
  • FIG. 2 is a graph showing the survival rate of mouse iPS cells when various ribonucleosides (uridine diphosphate (UDP) or uridine diphosphate-N-acetylglucosamine (UDP-GlcNAc)) were added in the presence of 10 ⁇ M BRQ.
  • 6 is a graph showing the survival rate of mouse ES cells when various ribonucleosides (UDP or UDP-GlcNAc) were added in the presence of 10 ⁇ M BRQ.
  • FIG. 6 is a graph showing the dose-dependent effect of nucleotide diphosphate on BRQ-dependent cytotoxicity in pluripotent stem cells.
  • ESC indicates the effect on mouse ES cells
  • iPSC indicates the effect on mouse iPS cells.
  • the circles show the dose-dependent effects of UDP, the diamonds of CDP, the squares of ADP, and the triangles of GDP. Error bars indicate ⁇ SD (standard deviation). Statistical significance was determined by t-test. ***P ⁇ 0.01, ***p ⁇ 0.001. It is a graph which shows the ratio of BrdU positive cell in a pluripotent stem cell in presence of BRQ.
  • ESC indicates the ratio in mouse ES cells
  • iPSC indicates the ratio in mouse iPS cell cells.
  • N and BRQ indicate the absence and presence of BRQ in the culture medium of pluripotent stem cells, respectively. It is a graph which shows the ratio of the Casp3 positive cell in a pluripotent stem cell in the presence of BRQ.
  • ESC indicates the ratio in mouse ES cells
  • iPSC indicates the ratio in mouse iPS cell cells.
  • N and BRQ indicate the absence and presence of BRQ in the culture medium of pluripotent stem cells, respectively. It is a photograph which shows the result of having analyzed the knockdown efficiency by a DHODH expression vector by western blotting.
  • the FLAG-tagged mouse DHODH expression vector was transferred to a Cos7 cell together with a vector encoding a control shRNA (“C” in the figure) or a vector encoding shRNA1 to 3 for DHODH (“DHODH sh1, sh2, sh3” in the figure). Introduced. Two days after the gene transfer, the cell extract was collected and analyzed by Western blotting using anti-FLAG antibody and anti-GAPDH antibody (loading control). It is a graph which shows the ratio of Ki67 positive cell in a control shRNA expression pluripotent stem cell or a DHODHshRNA expression pluripotent stem cell.
  • ESC and iPSC indicate the above-mentioned proportions in mouse ES cells and mouse iPS cells, respectively. Error bars indicate ⁇ SD. Statistical significance was determined by t-test. *P ⁇ 0.05, **P ⁇ 0.01, ***P ⁇ 0.001. It is a graph which shows the ratio of a Casp3 positive cell in a control shRNA expression pluripotent stem cell or a DHODHshRNA expression pluripotent stem cell. In the figure, “ESC” and “iPSC” indicate the above-mentioned proportions in mouse ES cells and mouse iPS cells, respectively. Error bars indicate ⁇ SD. Statistical significance was determined by t-test.
  • FIG. 3 is a fluorescence micrograph showing representative results obtained by observing control shRNA-expressing pluripotent stem cells and dhodhsh-expressing pluripotent stem cells by immunostaining for GFP and Ki67.
  • ESC and “iPSC” indicate the results of observing mouse ES cells and mouse iPS cells, respectively.
  • Each of the three photographs in the first and third rows shows the results of detecting Ki67 (red) expression and nuclei (blue, counterstaining with DAPI) in mouse ES cells and mouse iPS cells.
  • FIG. 3 is a fluorescence micrograph showing representative results obtained by observing control shRNA-expressing pluripotent stem cells and dhodhsh-expressing pluripotent stem cells by immunostaining for GFP and Casp3.
  • ESC and “iPSC” indicate the results of observing mouse ES cells and mouse iPS cells, respectively.
  • the three photographs in the first and third rows show the results of detecting the expression of Casp3 (red) and the nucleus (blue, counterstaining with DAPI) of mouse ES cells and mouse iPS cells.
  • the three photographs in the second and fourth rows show the results of detecting the expression of GFP (green) and Casp3 (red) in mouse ES cells and mouse iPS cells.
  • the scale bar shows 100 ⁇ m. After culturing for 24 hours in the presence of 10 ⁇ M BRQ, it is a photograph showing the results of observing each cell with a fluorescence microscope.
  • “NSC”, “iPS” and “ES” indicate the results of observing mouse neural stem cells, mouse iPS cells and mouse ES cells.
  • BRQ indicates the result of culturing in the presence of 10 ⁇ M BRQ
  • DMSO indicates the result of culturing in the absence of BRQ.
  • the upper 6 photographs show the results of immunostaining Sox2, and the lower 6 photographs are the photographs in which the results of the immunostaining of Sox2 and the results of counterstaining the cell nuclei with DAPI are superimposed.
  • Arrows indicate Sox2-negative cells. Each numerical value shows the ratio (%) of Sox2-positive cells to DAPI-positive cells. Scale bar represents 100 ⁇ m.
  • 3 is a graph showing the ratio (%) of Sox2-positive cells in each cell after culturing for 24 hours in the presence of 10 ⁇ M BRQ.
  • NSC NSC
  • iPS ES
  • ES ES
  • BRQ indicates the result of culturing in the presence of 10 ⁇ M BRQ
  • Const indicates the result of culturing in the absence of BRQ.
  • ** indicates that P value ⁇ 0.01.
  • 3 is a graph showing the expression levels of pluripotent stem cell marker genes (Sox2, Oct4 and Nanog) in each cell after culturing in the presence of 10 ⁇ M BRQ for 1 day or 2 days.
  • BRQ indicates the result of culturing in the presence of 10 ⁇ M BRQ
  • DMSO indicates the result of culturing in the absence of BRQ.
  • the vertical axis of each graph shows a relative value when the expression level of each gene in "DMSO” is 100.
  • ES and “iPS” indicate the results of observing mouse ES cells and mouse iPS cells.
  • BRQ indicates the result of culturing in the presence of 10 ⁇ M BRQ
  • cont indicates the result of culturing in the absence of BRQ.
  • the upper four photographs show the results of immunostaining for Nanog, and the lower four photographs are the photographs in which the results of immunostaining of Nanog and the results of counterstaining cell nuclei with DAPI are superimposed.
  • Arrows indicate cells in which Nanog is spread and distributed in the cytoplasm (cells in which Nanog is excluded from the nucleus).
  • Pluripotent stem cells were cultured for 2 days in the presence of DMSO, ZVAD (100 nM), BRQ (10 ⁇ M) or BRQ and ZVAD (BRQ:10 ⁇ M, ZVAD:100 nM), and immunolabeled with Nanog and Oct4. It is a fluorescence micrograph which shows a result.
  • ESC and “iPSC” indicate the results of observing mouse ES cells and mouse iPS cells.
  • the numerical values in the figure show the ratio of the number of cells positive for each nuclear PSC marker in all cells.
  • the upper 8 photographs show the results of immunostaining Oct4 (green), the middle 8 photographs show the results of immunostaining Nanog (red), and the lower 8 photographs show the results of immunostaining Oct4 and The result of superimposing the result of counter-staining the cell nucleus (blue) with DAPI on the result of immunostaining Nanog is shown.
  • the scale bar shows 50 ⁇ m.
  • Pluripotent stem cells were cultured in the presence of DMSO, BRQ (10 ⁇ M) or BRQ and LMB (BRQ:10 ⁇ M, LMB: 0.15 nM) for 2 days, and Nanog (red) and Oct4 (green) were cultured. It is a fluorescence micrograph which shows the result of immunolabeling.
  • ESC and "iPSC” indicate the results of observing mouse ES cells and mouse iPS cells, respectively.
  • the numerical values in the figure show the ratio of the number of cells positive for each nuclear PSC marker in all cells.
  • the upper 6 photographs show the results of immunostaining Oct4 (green), the middle 6 photographs show the results of immunostaining Nanog (red), and the lower 6 photographs show the results of immunostaining Oct4 and The results of immunostaining Nanog with the results of counterstaining the cell nuclei (blue) with DAPI are shown.
  • the scale bar shows 100 ⁇ m. Representative photographs of teratomas formed from pluripotent stem cells pretreated with DMSO or BRQ are shown. In the figure, “ESC” and “iPSC” indicate the results of observing teratomas formed from mouse ES cells and mouse iPS cells, respectively. Scale bar indicates 1 cm.
  • FIG. 6 is a graph showing the volume of tumors formed from pluripotent stem cells pretreated with DMSO or BRQ.
  • ESC and “iPSC” indicate the volume of tumor formed from mouse ES cells and mouse iPS cells, respectively. Error bars indicate ⁇ SD. Statistical significance was determined by t-test. ***P ⁇ 0.01, ***P ⁇ 0.001. Representative photographs of teratomas formed from pluripotent stem cells in mice treated with DMSO or BRQ are shown.
  • “ESC” and “iPSC” indicate the results of observing teratomas formed from mouse ES cells and mouse iPS cells, respectively. Scale bar indicates 1 cm.
  • 6 is a graph showing the tumor volume of teratomas formed from pluripotent stem cells in mice treated with DMSO or BRQ.
  • “ESC” and “iPSC” indicate tumor volumes formed from mouse ES cells and mouse iPS cells, respectively. Error bars indicate ⁇ SD.
  • Statistical significance was determined by t-test. ***P ⁇ 0.01, ***P ⁇ 0.001.
  • 6 is a graph showing the proportion of Ki67-positive cells in teratomas formed from pluripotent stem cells in mice treated with DMSO or BRQ.
  • "ESC” and “iPSC” indicate the above-mentioned proportions in teratoma formed from mouse ES cells and mouse iPS cells, respectively. Error bars indicate ⁇ SD.
  • ESC and “iPSC” indicate the results of observing teratomas formed from mouse ES cells and mouse iPS cells, respectively. Nuclei were detected by counterstaining with DAPI (blue). The scale bar shows 100 ⁇ m.
  • pluripotent stem cells can be cytotoxicized and removed. Revealed.
  • somatic stem cells obtained by inducing differentiation of pluripotent stem cells or differentiated cells such as somatic cells, even if DHODH is inhibited, significant cytotoxicity does not occur. ..
  • the present invention provides a composition and a method for removing undifferentiated pluripotent stem cells remaining in a cell population differentiated from pluripotent stem cells using a DHODH inhibitor.
  • dihydroorotic acid dehydrogenase means an enzyme that catalyzes the oxidation of dihydroorotic acid to orotic acid, which is the fourth reaction in the de novo synthetic pathway of pyrimidine, and includes, for example, dihydroorotic acid dehydrogenase (fumaric acid). ) (EC number: 1.3.98.1), dihydroorotic acid dehydrogenase (NAD+) (EC number: 1.3.1.14), dihydroorotic acid dehydrogenase (NADP+) (EC number: 1.3.1.15). ), and dihydroorotic acid dehydrogenase (quinone) (EC number: 1.3.5.2).
  • DHODH dihydroorotic acid dehydrogenase
  • DHODH inhibitor means a compound having an activity of inhibiting the catalytic reaction.
  • the “inhibition” in the present invention includes not only complete inhibition of activity and the like but also partial inhibition (suppression).
  • Examples of the “DHODH inhibitor” according to the present invention include brequinar (BRQ, 6-fluoro-2-(2′-fluoro-1,1′-biphenyl-4-yl), as shown in Examples below.
  • -3-methyl-4-quinoline-carboxylic acid sodium salt leflunomide (leflunomide, 5-methyl-N-[4-(trifluoromethyl)phenyl]-isoxazole-4-carboxamide), teriflunomide, (2Z )-2-Cyano-3-hydroxy-N-[4-(trifluoromethyl)phenyl]but-2-enamide), Vidofludimus, 2-(3-Fluoro-3′-methoxybiphenyl-4-ylcarbamoyl) )-Cyclopent-1-enecarboxylic acid
  • the “DHODH inhibitor” according to the present invention is preferably a compound having a 50% inhibitory concentration (IC 50 ) for human-derived DHODH of 100 nM or less, more preferably a compound having an IC 50 of 50 nM or less, and an IC 50 of 20 nM.
  • IC 50 50% inhibitory concentration
  • the following compounds are more preferable. Examples of such compounds include the following compounds (the structure of each compound and the IC 50 for human-derived DHODH are shown below).
  • the enzyme activity of DHODH can be determined by those skilled in the art by, for example, a dye reduction assay using dichloroindophenol (DCIP).
  • DCIP dichloroindophenol
  • the activity of DHODH can be determined using the decrease in the absorbance at the wavelength as an index.
  • the IC 50 for DHODH is, for example, that DOHDH derived from human, a substrate (dihydroorotic acid), an electron acceptor (for example, CoQ), and an enzyme reaction solution containing a buffer have various concentrations of a test compound (DHODH inhibition). Agent) and perform the DCIP assay (see WO 2008/077639, Biochem J. 1998 Dec 1;336(Pt2):299-303).
  • the “DHODH inhibitor” according to the present invention also includes a pharmacologically acceptable salt, hydrate or solvate as long as it has the inhibitory activity.
  • a pharmacologically acceptable salt is not particularly limited and can be appropriately selected according to each structure of the drug, and examples thereof include acid addition salts (hydrochloride, sulfate, hydrobromide). , Nitrates, hydrogensulfates, phosphates, acetates, etc.) and base addition salts (sodium salts, potassium salts, zinc salts, calcium salts, etc.).
  • the hydrate or solvate is not particularly limited, and examples thereof include those in which 0.1 to 10 molecules of water or solvent are added to 1 molecule of DHODH inhibitor or its salt.
  • the “DHODH inhibitor” includes all isomers such as tautomers, geometric isomers, optical isomers based on asymmetric carbons, stereoisomers and the like, as long as it has the inhibitory activity. Includes isomer mixtures.
  • DHODH inhibitors undergo metabolism in vivo such as oxidation, reduction, hydrolysis, amination, deamination, hydroxylation, phosphorylation, dehydroxylation, alkylation, dealkylation, conjugation, etc., and still inhibit the metabolism.
  • the present invention also includes compounds that exhibit activity, and the present invention also includes compounds that are metabolized in vivo by oxidation, reduction, hydrolysis and the like to produce DHODH inhibitors.
  • DHODH inhibitor many compounds have been developed and are on the market as described above, so they can be obtained by purchasing. Many reports have been made on methods for producing these compounds even if they are not commercially available. Therefore, a person skilled in the art can also appropriately prepare according to the manufacturing method.
  • the “pluripotent stem cell” may be any cell having pluripotency and self-renewal ability, and examples thereof include embryonic stem cells (ES cells), embryonal tumor cells (EC cells), and epiblasts. Stem cells (EpiS cells), embryonic germ cells (EG cells), pluripotent germ cells (mGS cells), MUSE cells (Kuroda Y. et al., Proc. Natl. Acad. Sci. USA, 2010) , Vol. 107, No. 19, pp. 8639-8643). Furthermore, "pluripotent stem cells” also include cells derived from somatic cells collected from a living body so as to have artificial pluripotency, such as induced pluripotent stem cells (iPS cells). Be done.
  • iPS cells induced pluripotent stem cells
  • the origin of these cells is not particularly limited, and humans and non-human animals (for example, rodents such as mice and rats, cows, horses, pigs, sheep, monkeys, dogs, mammals such as cats, chickens, etc.) Birds). Furthermore, in the case of aiming at the regenerative medicine described below, pluripotent stem cells derived from the target to which the differentiated cells described below are transplanted are desirable from the viewpoint of suppressing immune rejection.
  • undifferentiated pluripotent stem cells include, in addition to the pluripotent stem cells that have not been differentiated and maintain pluripotency, cells in which pluripotent stem cells have turned into tumors (for example, Teratoma, tetracarcinoma) are also included.
  • the “cell group differentiated from pluripotent stem cells” means a population of cells including any differentiated cells derived from the above pluripotent stem cells.
  • Such a cell group may include a single type of differentiated cell or may include a plurality of types of differentiated cells. Further, the form may be a tissue or an organ composed of these cells.
  • differentiated cells include not only cells that have completely lost pluripotency (terminally differentiated cells, such as somatic cells and germ cells) but also cells that have partially lost pluripotency (eg, progenitor cells). Cells, somatic stem cells) are also included.
  • somatic stem cells are cells also called adult stem cells and tissue stem cells, and include, for example, neural stem cells, satellite cells, hematopoietic stem cells (bone marrow stem cells), mesenchymal stem cells, intestinal stem cells, hair follicle stem cells, mammary stem cells, Examples include endothelial stem cells, olfactory mucosa stem cells, neural crest stem cells, and testis cells.
  • Induction of differentiation of pluripotent stem cells into “differentiated cells” is usually carried out through induction of differentiation into germ layers (ectodermal, mesoderm, endoderm) in which each differentiated cell develops.
  • a known method using a low-molecular compound, a protein or the like (differentiation inducing factor) suitable for inducing differentiation into each germ layer cell can be appropriately selected and performed.
  • ectodermal cells such as nerve cells can be induced to differentiate by culturing in the presence of a BMP inhibitor, TGF ⁇ and an activin inhibitor (Nat Biotechnol. 2009 Mar; 27(3):275. -80).
  • Differentiation can be induced into mesodermal cells such as chondrocytes by culturing pluripotent stem cells in the presence of Wnt and activin, and then culturing them in the presence of BMP4 ( Development.2008 Sep;135(17):2969-79).
  • endodermal cells such as pancreatic cells and intestinal cells can be induced to differentiate by culturing pluripotent stem cells in the presence of activin and then culturing them in the presence of BMP4 (for example, , Diabetes 2010 Sep; 59(9):2094-2101, Nature. 2011 Feb 3; 470(7332): 105-9).
  • “removal” of undifferentiated pluripotent stem cells remaining in the above-mentioned cell group means not only complete removal of the cells but also reduction of the number of the cells or the ratio in the cell group. To do.
  • the removal may be performed in vitro, or may be performed in vivo or ex vivo.
  • Embodiments of the present invention include a composition containing a DHODH inhibitor as an active ingredient for removing undifferentiated pluripotent stem cells remaining in a cell group differentiated from pluripotent stem cells.
  • composition of the present invention may contain a physiologically acceptable carrier in addition to the above-mentioned DHODH inhibitor.
  • physiologically acceptable carriers include physiological isotonic solutions (physiological saline, culture medium, glucose and other isotonic solutions containing D-sorbitol, D-mannitol, sodium chloride, etc.). ), excipients, preservatives, stabilizers (human serum albumin, polyethylene glycol, etc.), binders, solubilizing agents, nonionic surfactants, buffers (phosphate buffer, sodium acetate buffer, etc.) , Preservatives and antioxidants.
  • composition of the present invention may be in the form of a reagent used for research purposes or medical purposes (for example, production of differentiated cells, particularly differentiated cells with reduced tumorigenicity risk). Further, as described below, it may be in the form of a pharmaceutical composition to be administered to a subject into which the differentiated cells have been transplanted.
  • the present invention also provides a kit for producing a differentiated cell, particularly a differentiated cell with reduced tumorigenicity risk.
  • a kit for producing a differentiated cell in addition to the composition, pluripotent stem cells, the above-mentioned differentiation-inducing factor, differentiation-inducing medium, incubator, and other materials necessary for inducing differentiation of pluripotent stem cells into desired cells are included. be able to.
  • a reagent for detecting the cell-specific marker molecule for example, an antibody specific to the marker molecule
  • the kit of the present invention also includes instructions indicating the method of using the composition of the present invention, the method of inducing differentiation and the like.
  • the embodiment of the present invention also includes a step of contacting a group of cells differentiated from pluripotent stem cells with a dihydroorotic acid dehydrogenase inhibitor, a method of removing undifferentiated pluripotent stem cells remaining from the group of cells Are listed.
  • the “contact” between the above-mentioned DHODH inhibitor and the cell group can be performed by adding the DHODH inhibitor to the medium for maintaining the cell group.
  • the concentration to be added at that time is not particularly limited and can be appropriately adjusted by those skilled in the art depending on the types of the pluripotent stem cells and cell groups of interest, the type of DHODH inhibitor to be used, etc. It is 0.1 to 1000 ⁇ M, preferably 1 to 100 ⁇ M, and more preferably 10 to 50 ⁇ M.
  • the method of the present invention can be rephrased as "a method for producing differentiated cells with reduced tumorigenicity risk, which comprises the step of contacting a cell group differentiated from pluripotent stem cells with a dihydroorotate dehydrogenase inhibitor"
  • the DHODH inhibitor and the cell group can be treated by administering the DHODH inhibitor to the subject. Contact can be made.
  • the dosage form is not particularly limited, and examples thereof include intravenous administration, intraarterial administration, intraperitoneal administration, subcutaneous administration, intradermal administration, respiratory tract administration, rectal administration and intramuscular administration, administration by infusion, local administration, and oral administration. It is appropriately selected depending on the type of cell group to be transplanted, the site of transplantation and the like.
  • the dose is appropriately adjusted depending on the type of subject, age, weight, symptoms and health condition, administration form, types of target pluripotent stem cells and cell groups, types of DHODH inhibitors used, and the like.
  • the dose per administration is usually 0.001 to 1000 mg/kg body weight, preferably 0.1 to 100 mg/kg body weight, and more preferably 1 to 50 mg/kg body weight.
  • the number of administrations per day is also not particularly limited, and may be adjusted appropriately in consideration of various factors as described above. Further, the subject may be continuously administered (eg, once a week).
  • the contact time between the DHODH inhibitor and the cell population for removing the undifferentiated pluripotent stem cells may be any time sufficient for the removal, and those skilled in the art can appropriately adjust the time. However, it is usually 2 to 7 days, preferably 2 to 5 days, and more preferably 2 to 3 days.
  • mice All experiments using mice were performed according to the protocol approved by the Hokkaido University Animal Experiment Committee. The mouse used was purchased from Hokudo Co., Ltd. Unless otherwise specified, chemicals and growth factors used were those purchased from Invitrogen and PeproTech, respectively.
  • mice Pluripotent stem cells
  • E14tg2a a cell line given by Mr. Shinji Masui (at the time of receiving the cells, enrolled in Kyoto University iPS Laboratory) was used.
  • a cell line iPS-Hep-FB/Ng/gfp-103c-1 purchased from RIKEN BioResource Research Center was used.
  • pluripotent stem cells were cultured by the method disclosed on the SIGMA website (https://www.sigmaaldrich.com/life-science/stem-cell-biology/system-cell-protocols.html). reference).
  • a culture medium (ESC culture medium) having the following composition was used.
  • the surface of a 100 mm culture dish (manufactured by Falcon) was coated with gelatin derived from bovine epidermis (manufactured by Sigma, product number: G9391) and kept at 37° C. for 2 hours or more. The placed one was used.
  • EC cells Human embryonal carcinoma (EC) cells (NT2) were obtained from the American Tissue Culture Collection (CRL-1973) and cultured in DMEM (Dulbecco's modified Eagle medium) containing 10% FBS.
  • DMEM Dulbecco's modified Eagle medium
  • mice Mouse neural stem cells (mNSCs) are described in Johe KK et al., Genes Dev. 1996, Vol. 10, No. 24, pp. 3129-3140, prepared from the telencephalon of mice (fetus 14.5 days), and cultured in NSC medium [bFGF (10 ng/ml), EGF (10 ng/ml). ), heparin (5 ⁇ M), GlutaMAX (manufactured by Gibco), penicillin, and streptomycin in DMEM/F12 medium (manufactured by Sigma)].
  • Mouse bone marrow-derived stromal cells were obtained from Riken BioResource Center (RBRC-RCB1127) and cultured in ⁇ MEM containing 10% FBS (Eagle minimum essential medium ⁇ modified type).
  • Mouse myoblasts (C2C12) were obtained from Riken BioResource Center (RBRC-RCB0987) and cultured in DMEM containing 10% FBS.
  • Astrocytes were prepared by culturing mNSC in the presence of DMEM containing 10% FBS for 3 days, and then maintained in the same medium.
  • MTT assay method 1000 cells were seeded in each well of a 96-well plate and DMSO alone was added to a medium (200 ⁇ L/well) to a final concentration of 0.1%, or each dihydroorotic acid dehydrogenase (DHODH) inhibitor The cells were cultured for 3 days in a medium (200 ⁇ L/well) supplemented with.
  • the culture medium used was a culture medium suitable for each of the cells described above.
  • a DHODH inhibitor As a DHODH inhibitor, Brequnar (BRQ, Cayman Chemical, CAS number: 96187-53-0), Leflunomide (Cayman Chemical, CAS number: 75706-12-6), Teriflunomide (Cayman45:Chamical 81ChemicalChemical number: 81:Chamman: 45-81-Chemical number: 45-81-Chemical number: 16-Chemical-Chemical number) ) Or Vidofludimus (Cayman, CAS number: 717824-30-1) was added to the medium. The concentration of each DHODH inhibitor added to the medium was a 2-fold dilution series starting from 200 ⁇ M.
  • a well containing only the medium was prepared, and as a control, a well containing a medium containing Triton X-100 (final concentration 0.2%) was added to the cells.
  • ES cells or iPS cells were incubated for 3 days in the presence of BRQ (10 ⁇ M) or in the presence of BRQ (10 ⁇ M) and various concentrations of ribonucleosides (up to 500 ⁇ M, all manufactured by Sigma), and the MTT described above was added. Cell viability was analyzed by assay.
  • the cells were washed once with PBS, added with 4% paraformaldehyde, and incubated at room temperature for 10 minutes for fixation. After replacing with PBS containing 0.3% Triton X100 and incubating at room temperature for 5 minutes, it was replaced with PBS containing 0.3% Triton X100 and 10% FCS (blocking solution) and incubated at room temperature for 30 minutes. The solution was replaced with a primary antibody solution diluted with a blocking solution, and incubated at room temperature for 2 hours. After washing three times with PBS, the secondary antibody diluted with the blocking solution and the DAPI solution were added, and the mixture was incubated at room temperature for 2 hours.
  • the cells were encapsulated with an anti-fading agent (manufactured by DAKO, code number: S3023) and observed under a microscope.
  • the cell nuclei were visualized by counterstaining with a DAPI solution (1 ⁇ g/mL, manufactured by Dojindo, product code: D523).
  • the fluorescence image was acquired with an AxioImager M1 microscope (Carl Zeiss).
  • Anti-Nestin mouse monoclonal antibody (BD, clone Rat401, diluted 1:200 for use), Anti-Nanog rabbit polyclonal antibody (manufactured by Wako pure chemical, code number: 018-27521, used by diluting 1:200), Anti-Sox2 rabbit polyclonal antibody (manufactured by StemCell Technology, diluted 1:500 for use), Anti-Oct4 mouse monoclonal antibody (Cell Signaling Tech, diluted 1:200 for use), Anti-GFP rat monoclonal antibody (manufactured by Nacalai Tesque, Inc., diluted 1:500), Anti-Ki67 rabbit monoclonal antibody (Thermo Fisher Scientific, manufactured at a dilution of 1:100), Anti-active Caspase3 rabbit polyclonal antibody (manufactured by StemCell Technology,
  • Alexa488-labeled anti-mouse IgG donkey polyclonal antibody (Thermo Fisher Scientific, catalog number: R37120, diluted 1:500 for use), Alexa488-labeled anti-rabbit IgG donkey polyclonal antibody (Thermo Fisher Scientific, catalog number: A-11008, diluted 1:500 for use), Alexa 594-labeled anti-rabbit IgG donkey polyclonal antibody (Thermo Fisher Scientific, catalog number: R37119, diluted 1:500 for use), Alexa 594-labeled anti-mouse IgG donkey polyclonal antibody (Thermo Fisher Scientific, catalog number: R37115, diluted 1:500 for use), Alexa488 labeled anti-rat IgG goat antibody (manufactured by Jackson ImmunoResearch, diluted 1:500 and used).
  • Anti-Oct4 mouse monoclonal antibody Cell Signaling Tech, diluted 1:200 for use
  • Anti-Nanog rabbit polyclonal antibody manufactured by Wako Pure Chemical, diluted 1:200 for use
  • Anti-Ki67 rabbit monoclonal antibody Thermo Fisher Scientific, manufactured at a dilution of 1:100
  • Anti- ⁇ III tubulin mouse monoclonal antibody Sigma, diluted 1:400 for use
  • Anti-smooth muscle actin rabbit polyclonal antibody manufactured by Proteintech, SMA, diluted 1:200 for use
  • Anti-alpha fetoprotein rabbit polyclonal antibody Proteintech, AFP, diluted 1:100 for use).
  • Alexa 594-labeled anti-mouse IgG donkey polyclonal antibody (Thermo Fisher Scientific, catalog number: R37115, diluted 1:500 for use), Alexa488-labeled anti-rabbit IgG goat antibody (Jackson ImmunoResearch, 1:500 dilution).
  • mice NSCs and mouse ES cells or mouse iPS cells were seeded, and cultured in the ESC culture medium for 3 days in the presence of 1 ⁇ M or 10 ⁇ M BRQ. And it analyzed by the above-mentioned immunostaining.
  • Sox2 gene Forward primer 5'-TGAAGAAGGAATAAGTACACCGCT-3' (SEQ ID NO: 1), Reverse primer 5'-TCCTGCATCATGCTGTAGCTG-3' (SEQ ID NO: 2), oct4 gene: Forward primer 5′-CTGAAGCAGAAGAGGATCACC-3′ (SEQ ID NO: 3), Reverse primer 5'-CCGCAGCTTACACATGTTCTT-3' (SEQ ID NO: 4), nanog gene: Forward primer 5'-CTGATTCTTCTACCAGTCCCCAA-3' (SEQ ID NO: 5), Reverse primer 5'-AGAGTTCTTGCATCTGCTGGA-3' (SEQ ID NO: 6), 18S ribosomal RNA gene: Forward primer 5′-CGGACAGGATTGACAGATTG-3′ (SEQ ID NO: 7), Reverse primer 5'-CAAATCGCTCCACCAACTAA-3' (SEQ ID NO: 8). The expression level of each target gene thus detected was normalized by the relative quantification method (delt
  • Vectors are based on Nishide K, et al., PLoS ONE. 2009; 4(8):e6869. And Takanaga H, et al., Stem Cells. 2009;27(1):165-174. It was constructed according to the description.
  • the full-length cDNA of mouse DHODH is KOD Plus-Ver. 2 polymerase (manufactured by Toyobo Co., Ltd.) was used and amplified from mouse NSC-derived cDNA according to the instruction manual.
  • a 5'primer (5'-AGAATTCAATGGGGGAACA-3' (SEQ ID NO: 9)
  • a 3'primer (5'-TTGGATTCTCTGCCGGTC-3' (SEQ ID NO: 10) were used for the amplification.
  • the amplified cDNA was inserted into the p3xFLAG CMV10 vector to prepare p3xFLAG CMV10-mDHODH.
  • the target sequences of sh1 and sh3 targeting mouse DHODH are 5'-GGCTAGCTGTCtCTCTCT-3' (SEQ ID NO: 11) and 5'-GGAAGCTGTGTCTCTCA-3' (SEQ ID NO: 12), respectively.
  • the target sequence of sh(egfp) used as a control is 5'-GCAAGCTGACCCGTGTTTCA-3' (SEQ ID NO: 13).
  • the nucleotide sequence of the cloned cDNA was confirmed using BigDye terminator kit version 3.1 (manufactured by Applied Biosystems) and ABI sequencer model 3130xl (manufactured by Applied Biosystems).
  • Lipofectamine 3000 (Thermo Fisher Scientific Co., Ltd.) was used as a vector, and the vector was introduced into cells according to the instruction manual.
  • BRQ Brekiner sodium salt
  • Example 1 Examination on cytotoxic activity of DHODH inhibitor against pluripotent stem cells
  • Mouse ES cells and mouse iPS cells were cultured for 3 days in the presence of DHODH inhibitors (BRQ, Leflunomide, Teriflunomide or Videfludimus), respectively.
  • the survival rate was examined by the MTT assay.
  • Example 2 Examination on cytotoxic activity of DHODH inhibitor on somatic stem cells, etc.
  • pluripotent stem cells NT2: human embryonal carcinoma (EC) cells
  • PA6 mouse bone marrow-derived stromal cells
  • C2C12 mouse myoblasts
  • Example 3 Verification of cell-specific damaging activity of DHODH inhibitor on pluripotent stem cells
  • Pluripotent stem cells mouse iPS cells
  • Somatic stem cells mouse neural stem cells
  • ES cells and iPS cells disappeared in the presence of 10 ⁇ M BRQ, but no apparent cytotoxicity to neural stem cells was observed. I could't do it.
  • Example 4 Verification of Nucleoside Neutralizing Activity Against Cytotoxicity of DHODH Inhibitors Pyrimidine synthesis is regulated by nascent (de novo synthesis) and salvage (salvage) pathways. DHODH is a key factor in de novo synthesis that catalyzes the fourth rate-limiting chemical reaction in the synthetic pathway. Therefore, it was investigated whether the effect of removing pluripotent stem cells by the DHODH inhibitor can be eliminated by adding a nucleoside such as uridine which is a starting material of the salvage pathway.
  • a nucleoside such as uridine which is a starting material of the salvage pathway.
  • nucleoside adenosine, guanosine, cytidine or uridine
  • uridine a nucleoside
  • FIGS. 7 and 8 it was revealed that pyrimidine nucleosides (uridine and cytidine) have a neutralizing activity, and particularly uridine has a strong neutralizing activity against the cytotoxic activity of BRQ.
  • UDP-GlcNac uridine diphosphate-N-acetylglucosamine
  • OHT O-linked N-acetylglucosaminyltransferase
  • DHODH inhibitors such as BRQ exert cytotoxic activity on pluripotent stem cells by inhibiting the pyrimidine synthesis pathway. It was also suggested that its cytotoxic activity could be suppressed by activating the salvage pathway.
  • Example 6 Verification of Effect of DHODH Inhibitor on Marker Molecule of Pluripotent Stem Cell
  • three transcription factors Sox2 involved in promotion of self-renewal ability and maintenance of undifferentiated state.
  • Oct4, Nanog are expressed at high levels. Therefore, the effect of the DHODH inhibitor on the marker molecules of these pluripotent stem cells was examined.
  • Sox2 in pluripotent stem cells mouse iPS cells and mouse ES cells
  • somatic stem cells mouse neural stem cells
  • pluripotent stem cells mouse iPS cells and mouse ES cells
  • BRQ pluripotent stem cell marker gene
  • Nanog which is normally localized in the nucleus of pluripotent stem cells, becomes spread and distributed in the cytoplasm by BRQ treatment.
  • Example 7 Nuclear export of CRM1-dependent pluripotent stem cell marker molecule induced by DHODH inhibitor Pluripotent stem cells in the presence or absence of BRQ or pan-caspase inhibitor Z-VAD Were cultured. On the second day after treatment with BRQ, the number of Oct4-positive cells and Nanog-positive cells in pluripotent stem cells (ES cells and iPS cells) was measured.
  • Oct4 positive cells in ES cells treated with BRQ 2%, Nanog positive cells: 6%.
  • Oct4 positive cells 0%, Nanog positive cells: 0% in iPS cells treated with BRQ.
  • DMSO Oct4-positive cells: 85%, Nanog-positive cells: 86% in the ES cells treated with, and Oct4-positive cells: 94%, Nanog-positive cells: 93% in the iPS cells treated with DMSO).
  • pluripotent stem cells were cultured in a medium supplemented with leptomycin B (LMB) in the presence or absence of BRQ, and nuclear localization of Nanog and Oct4 was analyzed.
  • LMB is a specific inhibitor of CRM1 also known as Exportin1.
  • Example 8 Verification of suppressive effect of DHODH inhibitor on tumorigenicity of pluripotent stem cells
  • the tumorigenicity of pluripotent stem cells pretreated with BRQ which is a DHODH inhibitor was examined. Specifically, first, after culturing in a medium supplemented with DMSO alone or 10 ⁇ M BRQ for 2 days, surviving pluripotent stem cells were subcutaneously injected into the buttocks of the NOD/SCID mice. Then, 4 weeks after the transplantation, the tumor was excised and observed.
  • pluripotent stem cells were first transplanted subcutaneously into the buttocks of NOD/SCID mice to form tumors reaching a size of more than 100 mm 3 . Then, BRQ was intraperitoneally injected once every three days, and a tumor was excised from the mouse on day 3 after the fifth intraperitoneal injection.
  • BRQ administration suppressed tumor growth. Although not shown in the figure, no visible side effect was observed in the BRQ-administered mice. As also shown in FIG. 28, teratomas size treated with DMSO and BRQ is each a 0.55 cm 3 and 0.12 cm 3 in those from ES cells, those derived from iPS cells, 1.37Cm 3 and It was 0.32 cm 3 .
  • Ki67-positive proliferating cells in BRQ-treated tumors were significantly reduced as compared with those in DMSO-treated tumors.
  • the number of Oct4-positive cells and Nanog-positive cells was significantly reduced in BRQ-treated tumors, but many cells expressing pluripotent stem cell markers were observed in DMSO-treated tumors. It was On the other hand, both tumors similarly contained cells expressing ATF, ⁇ III tubulin and SMA, which are markers in the three germ layers, respectively.
  • the DHODH inhibitor can suppress tumor formation by specifically removing undifferentiated pluripotent stem cells without causing obvious cytotoxicity to differentiated cells and mice.
  • the present invention it is possible to remove undifferentiated pluripotent stem cells without causing significant cytotoxicity to the differentiated cells. Therefore, the present invention is excellent in removing undifferentiated pluripotent stem cells remaining in a cell group differentiated from pluripotent stem cells, so that the risk of tumorigenesis is reduced, and regenerative medicine with few side effects, etc. Is useful in.

Abstract

The purpose of the present invention is to provide a composition and method for removing undifferentiated pluripotent stem cells that remain present in a cell group that has been induced to differentiate from pluripotent stem cells. It has been discovered that a dihydroorotate dehydrogenase inhibitor exhibits cytotoxicity for pluripotent stem cells while not exhibiting significant cytotoxicity for cells that have undergone differentiation, e.g., somatic stem cells.

Description

多能性幹細胞を除去するための組成物、及び多能性幹細胞の除去方法Composition for removing pluripotent stem cells and method for removing pluripotent stem cells
 本発明は、多能性幹細胞を除去するための組成物、及び多能性幹細胞の除去方法に関する。より詳しくは、多能性幹細胞から分化誘導した細胞群に残存する未分化の多能性幹細胞を除去するための組成物及び方法に関する。 The present invention relates to a composition for removing pluripotent stem cells and a method for removing pluripotent stem cells. More specifically, the present invention relates to a composition and method for removing undifferentiated pluripotent stem cells remaining in a cell group in which differentiation is induced from pluripotent stem cells.
 胚性幹細胞(ES細胞)、人工多能性幹細胞(iPS細胞)等の多能性幹細胞は、生体を構成する全ての細胞に分化する能力を有することから再生医療実現化の要としてその利用が期待されている。そして、現在までに、これら多能性幹細胞から治療に必要とされる特定の移植用機能細胞への分化誘導法が多々確立されている。 Pluripotent stem cells such as embryonic stem cells (ES cells) and induced pluripotent stem cells (iPS cells) have the ability to differentiate into all the cells that make up the living body, and therefore they are used as the key to the realization of regenerative medicine. Is expected. In addition, until now, various methods for inducing differentiation from these pluripotent stem cells to specific functional cells for transplantation required for treatment have been established.
 しかしながら、分化誘導後に残存した未分化の多能性幹細胞は、腫瘍を形成する可能性がある。そのため、移植治療を進める上で大きな障害となっており、この問題を解決するため、多能性幹細胞を除去する様々な方法が開発されている(非特許文献1~5)。 However, undifferentiated pluripotent stem cells remaining after differentiation induction may form tumors. Therefore, it is a major obstacle to the promotion of transplantation therapy, and various methods for removing pluripotent stem cells have been developed to solve this problem (Non-Patent Documents 1 to 5).
 しかし、これらの方法による他の細胞への安全性についての検証は不十分なこともあり、副作用少なく、多能性幹細胞を除去する方法は未だ実用化されていないのが現状である。 However, the verification of the safety to other cells by these methods may be insufficient, there are few side effects, and the method of removing pluripotent stem cells has not yet been put to practical use.
 本発明は、前記従来技術の有する課題に鑑みてなされたものであり、未分化の多能性幹細胞に対して細胞傷害性をもたらす一方で、他の細胞に対しては毒性の少ない化合物を見出すことを目的とする。さらには、該化合物を有効成分とする多能性幹細胞を除去するための組成物、及び前記化合物を用いた多能性幹細胞の除去方法を提供することを、本発明の目的とする。 The present invention has been made in view of the above problems of the prior art, and finds a compound that brings cytotoxicity to undifferentiated pluripotent stem cells, while having little toxicity to other cells. The purpose is to Furthermore, it is an object of the present invention to provide a composition for removing pluripotent stem cells containing the compound as an active ingredient, and a method for removing pluripotent stem cells using the compound.
 本発明者は、前記目的を達成すべく鋭意研究を重ねた結果、ジヒドロオロト酸デヒドロゲナーゼ(DHODH)阻害剤が、ES細胞及びiPS細胞等の多能性幹細胞に対して細胞傷害活性を示すことを見出した。一方、DHODH阻害剤は、分化した細胞(アストロサイト等の体細胞、神経幹細胞等の体性幹細胞)に対しては、有意な細胞傷害性を示さないことを明らかにした。 The present inventors have conducted extensive studies to achieve the above-mentioned object, and as a result, found that a dihydroorotate dehydrogenase (DHODH) inhibitor shows cytotoxic activity against pluripotent stem cells such as ES cells and iPS cells. It was On the other hand, it was revealed that DHODH inhibitors do not show significant cytotoxicity to differentiated cells (somatic cells such as astrocytes and somatic stem cells such as neural stem cells).
 さらに、多能性幹細胞をDHODH阻害剤にて処理した上でマウスに移植することによって、または多能性幹細胞を移植したマウスにDHODH阻害剤を投与することによって、前記マウスに特段の副作用をもたらすことなく、多能性幹細胞からの腫瘍形成を抑制できることも確認した。 Furthermore, by treating pluripotent stem cells with a DHODH inhibitor and then transplanting them into a mouse, or by administering a DHODH inhibitor to a mouse into which a pluripotent stem cells are transplanted, a particular side effect is brought about in the mouse. It was also confirmed that the tumor formation from pluripotent stem cells could be suppressed without the above.
 DHODHは、ピリミジンのde novo合成の4番目の化学反応を触媒する酸化還元酵素である。DHODH阻害剤は、その合成を阻害することによって、T細胞やB細胞の増殖を抑制し、免疫抑制効果を発揮することが既に明らかとなっており、当該薬剤は、かかる効果故、関節リウマチ等の自己免疫疾患の治療に用いられている。さらに、DHODH阻害剤については、がん幹細胞における分化阻害を解除することによって、または同細胞におけるp53の合成を増強することによって、白血病(急性骨髄性白血病、慢性骨髄性白血病)に対する治療効果を奏することも報告されている(非特許文献6及び7)。 DHODH is an oxidoreductase that catalyzes the fourth chemical reaction of de novo synthesis of pyrimidine. It has already been clarified that the DHODH inhibitor suppresses the proliferation of T cells and B cells and exerts an immunosuppressive effect by inhibiting the synthesis thereof, and the drug is due to such an effect, rheumatoid arthritis and the like. Is used for the treatment of autoimmune diseases of. Furthermore, DHODH inhibitors exert a therapeutic effect on leukemia (acute myeloid leukemia, chronic myelogenous leukemia) by releasing differentiation inhibition in cancer stem cells or by enhancing p53 synthesis in the same cells. It has also been reported (Non-patent documents 6 and 7).
 しかしながら、DHODH阻害剤の多能性幹細胞への影響については何ら検証も示唆もされていなかったものの、本発明者は、上述のとおり、当該薬剤によって多能性幹細胞を除去できることを初めて明らかにした。その一方で、多能性幹細胞と比して劣るとはいえ多分化能と自己増殖性を備え、同じく幹細胞に分類される、体性幹細胞(神経幹細胞等)に対しては、DHODH阻害剤は有意な細胞傷害性を示さないことも、本発明者は見出し、本発明を完成するに至った。 However, although no verification or suggestion was made on the effect of the DHODH inhibitor on pluripotent stem cells, the present inventor revealed for the first time that the drug can remove pluripotent stem cells, as described above. .. On the other hand, for somatic stem cells (neural stem cells, etc.), which are inferior to pluripotent stem cells but have pluripotency and self-proliferative ability and are also classified as stem cells, a DHODH inhibitor is The present inventors have found that they do not show significant cytotoxicity, and have completed the present invention.
 すなわち、本発明は、DHODH阻害剤を用いた、多能性幹細胞を除去するための組成物、及び多能性幹細胞の除去方法に関し、より具体的には以下を提供する。
<1> ジヒドロオロト酸デヒドロゲナーゼ阻害剤を有効成分として含有する、多能性幹細胞から分化誘導した細胞群に残存する未分化の多能性幹細胞を除去するための組成物。
<2> 前記多能性幹細胞が、胚性幹細胞(ES細胞)、人工多能性幹細胞(iPS細胞)及び胚性腫瘍細胞(EC細胞)からなる群から選択される少なくとも1の多能性幹細胞である、<1>に記載の組成物。
<3> 前記ジヒドロオロト酸デヒドロゲナーゼ阻害剤がブレキナー(brequinar)である、<1>又は<2>に記載の組成物。
<4> 前記細胞群が、前記多能性幹細胞から分化誘導した体性幹細胞を含む細胞群である、<1>~<3>のうちのいずれか一項に記載の組成物。
<5> 多能性幹細胞から分化誘導した細胞群とジヒドロオロト酸デヒドロゲナーゼ阻害剤とを接触させる工程を含む、前記細胞群から残存する未分化の多能性幹細胞を除去する方法。
<6> 前記多能性幹細胞が、胚性幹細胞(ES細胞)、人工多能性幹細胞(iPS細胞)及び胚性腫瘍細胞(EC細胞)からなる群から選択される少なくとも1の多能性幹細胞である、<5>に記載の方法。
<7> 前記ジヒドロオロト酸デヒドロゲナーゼ阻害剤がブレキナー(brequinar)である、<5>又は<6>に記載の方法。
<8> 前記細胞群が、前記多能性幹細胞から分化誘導した体性幹細胞を含む細胞群である、<5>~<7>のうちのいずれか一項に記載の方法。
That is, the present invention relates to a composition for removing pluripotent stem cells and a method for removing pluripotent stem cells using a DHODH inhibitor, and more specifically provides the following.
<1> A composition for removing undifferentiated pluripotent stem cells remaining in a cell group differentiated from pluripotent stem cells, containing a dihydroorotic acid dehydrogenase inhibitor as an active ingredient.
<2> The pluripotent stem cell is at least one pluripotent stem cell selected from the group consisting of embryonic stem cells (ES cells), induced pluripotent stem cells (iPS cells) and embryonic tumor cells (EC cells) The composition according to <1>.
<3> The composition according to <1> or <2>, wherein the dihydroorotic acid dehydrogenase inhibitor is brequinar.
<4> The composition according to any one of <1> to <3>, wherein the cell group is a cell group containing somatic stem cells differentiated from the pluripotent stem cells.
<5> A method for removing residual undifferentiated pluripotent stem cells from the cell group, which comprises a step of contacting a cell group differentiated from the pluripotent stem cells with a dihydroorotate dehydrogenase inhibitor.
<6> The pluripotent stem cell is at least one pluripotent stem cell selected from the group consisting of embryonic stem cells (ES cells), induced pluripotent stem cells (iPS cells) and embryonic tumor cells (EC cells) The method according to <5>.
<7> The method according to <5> or <6>, wherein the dihydroorotic acid dehydrogenase inhibitor is brequinar.
<8> The method according to any one of <5> to <7>, wherein the cell group is a cell group containing somatic stem cells differentiated from the pluripotent stem cells.
 本発明によれば、分化細胞には有意な細胞傷害をもたらすことなく、未分化の多能性幹細胞を除去することが可能となる。すなわち、本発明によれば、多能性幹細胞から分化誘導した細胞群に残存する未分化の多能性幹細胞を除去することが可能となる。 According to the present invention, undifferentiated pluripotent stem cells can be removed without causing significant cytotoxicity to differentiated cells. That is, according to the present invention, it becomes possible to remove the undifferentiated pluripotent stem cells remaining in the cell group differentiated from the pluripotent stem cells.
ジヒドロオロト酸デヒドロゲナーゼ(DHODH)阻害剤存在下3日間培養した後の、マウス胚性幹(ES)細胞の生存率を示すグラフである。図中、「BRQ」、「Leflunomide]、「Teriflunomide」及び「Vidofludimus」は、各DHODH阻害剤(ブレキナー、レフルノミド、テリフルノミド及びビドフルジムス)存在下における細胞の生存率を示す(図中の表記については、図2においても同様である)。1 is a graph showing the survival rate of mouse embryonic stem (ES) cells after culturing for 3 days in the presence of a dihydroorotic acid dehydrogenase (DHODH) inhibitor. In the figure, “BRQ”, “Leflunomide”, “Teriflunomide” and “Vidofludimus” indicate the viability of cells in the presence of each DHODH inhibitor (Brekiner, Leflunomide, Teriflunomide and Bidofludimus) (for the notation in the figure, The same applies to FIG. 2). DHODH阻害剤存在下3日間培養した後の、マウス人工多能性幹(iPS)細胞の生存率を示すグラフである。It is a graph which shows the survival rate of mouse induced pluripotent stem (iPS) cells after 3 days of culture in the presence of a DHODH inhibitor. BRQ存在下3日間培養した後の、マウス神経幹細胞等の生存率を示すグラフである。図中、「mNSC」、「mNSC+5%FCS」及び「アストロサイト」は、BRQ存在下における、マウス神経幹細胞、5%ウシ胎仔血清添加時のマウス神経幹細胞及びマウスアストロサイトの生存率を各々示す。It is a graph which shows the survival rate of mouse neural stem cells etc. after 3 days of culture in the presence of BRQ. In the figure, “mNSC”, “mNSC+5% FCS”, and “astrocyte” represent the survival rates of mouse neural stem cells and mouse astrocytes in the presence of BRQ when 5% fetal bovine serum was added. 10μM BRQ存在下3日間培養後の、各種細胞の生存率を示すグラフである。図中、「ES」、「iPS」、「NT2」、「PA6」、「C2C12」、「NSC」及び「アストロサイト」は、マウスES細胞、マウスiPS細胞、ヒト胚性癌細胞、マウス骨髄由来ストローマ細胞、マウス筋芽細胞、マウス神経幹細胞及びマウスアストロサイトの前記生存率を各々示す。It is a graph showing the survival rate of various cells after 3 days of culture in the presence of 10 μM BRQ. In the figure, “ES”, “iPS”, “NT2”, “PA6”, “C2C12”, “NSC” and “astrosite” are derived from mouse ES cells, mouse iPS cells, human embryonal carcinoma cells and mouse bone marrow. The survival rates of stromal cells, mouse myoblasts, mouse neural stem cells and mouse astrocytes are shown respectively. BRQ存在下において、マウス神経幹細胞及びマウスES細胞を3日間混合培養した結果を示す、蛍光顕微鏡写真である。図中、「コントロール」はBRQ非存在下における培養結果を示す。図中、上段の3写真は、神経幹細胞のマーカー(Nestin)を免疫染色にて検出した結果を示す。中段の3写真は、多能性幹細胞のマーカー(Nanog)を免疫染色にて検出した結果を示す。下段の3写真は、Nestin及びNanogを免疫染色にて検出した結果と、DAPIによって細胞核を対比染色した結果とを重ね合わせた写真である。3 is a fluorescence micrograph showing the results of mixed culture of mouse neural stem cells and mouse ES cells for 3 days in the presence of BRQ. In the figure, "control" indicates the culture result in the absence of BRQ. In the figure, the upper three photographs show the results of detection of a neural stem cell marker (Nestin) by immunostaining. The middle three photographs show the results of detecting pluripotent stem cell markers (Nanog) by immunostaining. The bottom three photographs are photographs in which the results of detecting Nestin and Nanog by immunostaining and the results of counterstaining the cell nuclei with DAPI are superimposed. BRQ存在下において、マウス神経幹細胞及びマウスiPS細胞を3日間混合培養した結果を示す、蛍光顕微鏡写真である。図中、「コントロール」はBRQ非存在下における培養結果を示す。図中、上段の3写真は、多能性幹細胞のマーカー(Nanog)を免疫染色にて検出した結果を示す。中段の3写真は、神経幹細胞のマーカー(Nestin)を免疫染色にて検出した結果を示す。下段の3写真は、Nestin及びNanogを免疫染色にて検出した結果と、DAPIによって細胞核を対比染色した結果とを重ね合わせた写真である。3 is a fluorescence micrograph showing the results of mixed culture of mouse neural stem cells and mouse iPS cells for 3 days in the presence of BRQ. In the figure, "control" indicates the culture result in the absence of BRQ. In the figure, the upper three photographs show the results of detecting a pluripotent stem cell marker (Nanog) by immunostaining. The middle three photographs show the results of detecting a neural stem cell marker (Nestin) by immunostaining. The bottom three photographs are photographs in which the results of detecting Nestin and Nanog by immunostaining and the results of counterstaining the cell nuclei with DAPI are superimposed. 10μM BRQ存在下、各種リボヌクレオシド(ウリジン、アデノシン、グアノシン又はシチジン)を添加した際の、マウスiPS細胞の生存率を示すグラフである。9 is a graph showing the survival rate of mouse iPS cells when various ribonucleosides (uridine, adenosine, guanosine or cytidine) were added in the presence of 10 μM BRQ. 10μM BRQ存在下、各種リボヌクレオシド(ウリジン、アデノシン、グアノシン又はシチジン)を添加した際の、マウスES細胞の生存率を示すグラフである。6 is a graph showing the survival rate of mouse ES cells when various ribonucleosides (uridine, adenosine, guanosine or cytidine) were added in the presence of 10 μM BRQ. 10μM BRQ存在下、各種リボヌクレオシド(ウリジン二リン酸(UDP)又はウリジン二リン酸-N-アセチルグルコサミン(UDP-GlcNAc))を添加した際の、マウスiPS細胞の生存率を示すグラフである。2 is a graph showing the survival rate of mouse iPS cells when various ribonucleosides (uridine diphosphate (UDP) or uridine diphosphate-N-acetylglucosamine (UDP-GlcNAc)) were added in the presence of 10 μM BRQ. 10μM BRQ存在下、各種リボヌクレオシド(UDP又はUDP-GlcNAc)を添加した際の、マウスES細胞の生存率を示すグラフである。6 is a graph showing the survival rate of mouse ES cells when various ribonucleosides (UDP or UDP-GlcNAc) were added in the presence of 10 μM BRQ. 多能性幹細胞におけるBRQ依存的細胞傷害性に対する、ヌクレオチド二リン酸の用量依存効果を示すグラフである。図中、「ESC」はマウスES細胞に対する効果を示し、「iPSC」はマウスiPS細胞に対する効果を示す。また各グラフにおいて、丸はUDPの、ダイアモンドはCDPの、四角はADPの、三角はGDPの用量依存効果を示す。エラーバーは±SD(標準偏差)を示す。統計学的有意性はt検定によって判定した。**p<0.01、***p<0.001。FIG. 6 is a graph showing the dose-dependent effect of nucleotide diphosphate on BRQ-dependent cytotoxicity in pluripotent stem cells. In the figure, “ESC” indicates the effect on mouse ES cells, and “iPSC” indicates the effect on mouse iPS cells. In each graph, the circles show the dose-dependent effects of UDP, the diamonds of CDP, the squares of ADP, and the triangles of GDP. Error bars indicate ±SD (standard deviation). Statistical significance was determined by t-test. ***P<0.01, ***p<0.001. BRQ存在下での、多能性幹細胞におけるBrdU陽性細胞の割合を示すグラフである。図中、「ESC」はマウスES細胞における前記割合を示し、「iPSC」はマウスiPS細胞細胞における前記割合を示す。「N」及び「BRQ」は、多能性幹細胞の培養培地においてBRQが各々存在していない及び存在していることを示す。It is a graph which shows the ratio of BrdU positive cell in a pluripotent stem cell in presence of BRQ. In the figure, "ESC" indicates the ratio in mouse ES cells, and "iPSC" indicates the ratio in mouse iPS cell cells. "N" and "BRQ" indicate the absence and presence of BRQ in the culture medium of pluripotent stem cells, respectively. BRQ存在下での、多能性幹細胞におけるCasp3陽性細胞の割合を示すグラフである。図中、「ESC」はマウスES細胞における前記割合を示し、「iPSC」はマウスiPS細胞細胞における前記割合を示す。「N」及び「BRQ」は、多能性幹細胞の培養培地においてBRQが各々存在していない及び存在していることを示す。It is a graph which shows the ratio of the Casp3 positive cell in a pluripotent stem cell in the presence of BRQ. In the figure, "ESC" indicates the ratio in mouse ES cells, and "iPSC" indicates the ratio in mouse iPS cell cells. "N" and "BRQ" indicate the absence and presence of BRQ in the culture medium of pluripotent stem cells, respectively. DHODH発現ベクターによるノックダウン効率をウエスタンブロッティングにより分析した結果を示す写真である。FLAGタグ標識マウスDHODH発現ベクターを、コントロールshRNAをコードするベクター(図中「C」)又はDHODHに対するshRNA1~3を各々コードするベクター(図中、「DHODH sh1,sh2、sh3」)と共にCos7細胞に導入した。遺伝子導入してから2日後に、細胞抽出液を回収し、抗FLAG抗体及び抗GAPDH抗体(ローディングコントロール)を用いたウエスタンブロッティングによって分析した。It is a photograph which shows the result of having analyzed the knockdown efficiency by a DHODH expression vector by western blotting. The FLAG-tagged mouse DHODH expression vector was transferred to a Cos7 cell together with a vector encoding a control shRNA (“C” in the figure) or a vector encoding shRNA1 to 3 for DHODH (“DHODH sh1, sh2, sh3” in the figure). Introduced. Two days after the gene transfer, the cell extract was collected and analyzed by Western blotting using anti-FLAG antibody and anti-GAPDH antibody (loading control). コントロールshRNA発現多能性幹細胞又はDHODHshRNA発現多能性幹細胞における、Ki67陽性細胞の割合を示すグラフである。図中「ESC」及び「iPSC」は、マウスES細胞及びマウスiPS細胞各々における前記割合を示す。エラーバーは±SDを示す。統計学的有意性は、t検定によって判定した。*P<0.05,**P<0.01,***P<0.001。It is a graph which shows the ratio of Ki67 positive cell in a control shRNA expression pluripotent stem cell or a DHODHshRNA expression pluripotent stem cell. In the figure, "ESC" and "iPSC" indicate the above-mentioned proportions in mouse ES cells and mouse iPS cells, respectively. Error bars indicate ±SD. Statistical significance was determined by t-test. *P<0.05, **P<0.01, ***P<0.001. コントロールshRNA発現多能性幹細胞又はDHODHshRNA発現多能性幹細胞における、Casp3陽性細胞の割合を示すグラフである。図中「ESC」及び「iPSC」は、マウスES細胞及びマウスiPS細胞各々における前記割合を示す。エラーバーは±SDを示す。統計学的有意性は、t検定によって判定した。*P<0.05,**P<0.01,***P<0.001。It is a graph which shows the ratio of a Casp3 positive cell in a control shRNA expression pluripotent stem cell or a DHODHshRNA expression pluripotent stem cell. In the figure, "ESC" and "iPSC" indicate the above-mentioned proportions in mouse ES cells and mouse iPS cells, respectively. Error bars indicate ±SD. Statistical significance was determined by t-test. *P<0.05, **P<0.01, ***P<0.001. コントロールshRNA発現多能性幹細胞及びdhodh sh発現多能性幹細胞に、GFP及びKi67を対象とする免疫染色を施して観察し、得られた代表的な結果を示す、蛍光顕微鏡写真である。図中「ESC」及び「iPSC」は、マウスES細胞及びマウスiPS細胞各々を観察した結果を示す。1段目及び3段目における各3写真は、マウスES細胞及びマウスiPS細胞のKi67(赤色)の発現及び核(青色、DAPIによる対比染色)を検出した結果を示す。2段目及び4段目における各3写真は、マウスES細胞及びマウスiPS細胞のGFP(緑色)及びKi67(赤色)の発現を検出した結果を示す。スケールバーは100μmを示す。Fig. 3 is a fluorescence micrograph showing representative results obtained by observing control shRNA-expressing pluripotent stem cells and dhodhsh-expressing pluripotent stem cells by immunostaining for GFP and Ki67. In the figure, “ESC” and “iPSC” indicate the results of observing mouse ES cells and mouse iPS cells, respectively. Each of the three photographs in the first and third rows shows the results of detecting Ki67 (red) expression and nuclei (blue, counterstaining with DAPI) in mouse ES cells and mouse iPS cells. Each of the three photographs in the second and fourth rows shows the results of detecting the expression of GFP (green) and Ki67 (red) in mouse ES cells and mouse iPS cells. The scale bar shows 100 μm. コントロールshRNA発現多能性幹細胞及びdhodh sh発現多能性幹細胞に、GFP及びCasp3を対象とする免疫染色を施して観察し、得られた代表的な結果を示す、蛍光顕微鏡写真である。図中「ESC」及び「iPSC」は、マウスES細胞及びマウスiPS細胞各々を観察した結果を示す。1段目及び3段目における各3写真は、マウスES細胞及びマウスiPS細胞のCasp3(赤色)の発現及び核(青色、DAPIによる対比染色)を検出した結果を示す。2段目及び4段目における各3写真は、マウスES細胞及びマウスiPS細胞のGFP(緑色)及びCasp3(赤色)の発現を検出した結果を示す。スケールバーは100μmを示す。Fig. 3 is a fluorescence micrograph showing representative results obtained by observing control shRNA-expressing pluripotent stem cells and dhodhsh-expressing pluripotent stem cells by immunostaining for GFP and Casp3. In the figure, “ESC” and “iPSC” indicate the results of observing mouse ES cells and mouse iPS cells, respectively. The three photographs in the first and third rows show the results of detecting the expression of Casp3 (red) and the nucleus (blue, counterstaining with DAPI) of mouse ES cells and mouse iPS cells. The three photographs in the second and fourth rows show the results of detecting the expression of GFP (green) and Casp3 (red) in mouse ES cells and mouse iPS cells. The scale bar shows 100 μm. 10μM BRQ存在下で24時間培養した後に、各細胞を蛍光顕微鏡にて観察した結果を示す写真である。図中、「NSC」、「iPS」及び「ES」は、マウス神経幹細胞、マウスiPS細胞及びマウスES細胞を観察した結果を示す。「BRQ」は10μM BRQ存在下で培養した結果を示し、「DMSO」はBRQ非存在下で培養した結果を示す。上段の6写真はSox2を免疫染色した結果を示し、下段の6写真はSox2を免疫染色した結果とDAPIによって細胞核を対比染色した結果とを重ね合わせた写真である。矢印はSox2陰性細胞を示す。各数値はDAPI陽性細胞に対するSox2陽性細胞の割合(%)を示す。スケールバーは100μmを表す。After culturing for 24 hours in the presence of 10 μM BRQ, it is a photograph showing the results of observing each cell with a fluorescence microscope. In the figure, “NSC”, “iPS” and “ES” indicate the results of observing mouse neural stem cells, mouse iPS cells and mouse ES cells. “BRQ” indicates the result of culturing in the presence of 10 μM BRQ, and “DMSO” indicates the result of culturing in the absence of BRQ. The upper 6 photographs show the results of immunostaining Sox2, and the lower 6 photographs are the photographs in which the results of the immunostaining of Sox2 and the results of counterstaining the cell nuclei with DAPI are superimposed. Arrows indicate Sox2-negative cells. Each numerical value shows the ratio (%) of Sox2-positive cells to DAPI-positive cells. Scale bar represents 100 μm. 10μM BRQ存在下で24時間培養した後の各細胞における、Sox2陽性細胞の割合(%)を示すグラフである。図中、「NSC」、「iPS」及び「ES」は、マウス神経幹細胞、マウスiPS細胞及びマウスES細胞における各Sox2陽性細胞の割合を示す。「BRQ」は10μM BRQ存在下で培養した結果を示し、「Cont」はBRQ非存在下で培養した結果を示す。「**」はP値<0.01であることを示す。3 is a graph showing the ratio (%) of Sox2-positive cells in each cell after culturing for 24 hours in the presence of 10 μM BRQ. In the figure, “NSC”, “iPS” and “ES” indicate the proportion of each Sox2-positive cell in mouse neural stem cells, mouse iPS cells and mouse ES cells. “BRQ” indicates the result of culturing in the presence of 10 μM BRQ, and “Cont” indicates the result of culturing in the absence of BRQ. "**" indicates that P value <0.01. 10μM BRQ存在下で1日又は2日培養した後の、各細胞における多能性幹細胞マーカー遺伝子(Sox2、Oct4及びNanog)の発現量を示すグラフである。図中、「ES」及び「iPS」は、マウスES細胞及びマウスiPS細胞における各遺伝子の発現量を示す。「BRQ」は10μM BRQ存在下で培養した結果を示し、「DMSO」はBRQ非存在下で培養した結果を示す。各グラフの縦軸は、「DMSO」における各遺伝子の発現量を100とした場合の相対的な値を示す。3 is a graph showing the expression levels of pluripotent stem cell marker genes (Sox2, Oct4 and Nanog) in each cell after culturing in the presence of 10 μM BRQ for 1 day or 2 days. In the figure, “ES” and “iPS” indicate the expression level of each gene in mouse ES cells and mouse iPS cells. “BRQ” indicates the result of culturing in the presence of 10 μM BRQ, and “DMSO” indicates the result of culturing in the absence of BRQ. The vertical axis of each graph shows a relative value when the expression level of each gene in "DMSO" is 100. 10μM BRQ存在下で2日間培養した後に、各細胞を蛍光顕微鏡にて観察した結果を示す写真である。図中、「ES」及び「iPS」は、マウスES細胞及びマウスiPS細胞を観察した結果を示す。「BRQ」は10μM BRQ存在下で培養した結果を示し、「cont」はBRQ非存在下で培養した結果を示す。上段の4写真はNanogを免疫染色した結果を示し、下段の4写真はNanogを免疫染色した結果とDAPIによって細胞核を対比染色した結果とを重ね合わせた写真である。矢印はNanogが細胞質に広がって分布している細胞(Nanogが核外に排除された細胞)を示す。After culturing in the presence of 10 μM BRQ for 2 days, it is a photograph showing the results of observing each cell with a fluorescence microscope. In the figure, "ES" and "iPS" indicate the results of observing mouse ES cells and mouse iPS cells. “BRQ” indicates the result of culturing in the presence of 10 μM BRQ, and “cont” indicates the result of culturing in the absence of BRQ. The upper four photographs show the results of immunostaining for Nanog, and the lower four photographs are the photographs in which the results of immunostaining of Nanog and the results of counterstaining cell nuclei with DAPI are superimposed. Arrows indicate cells in which Nanog is spread and distributed in the cytoplasm (cells in which Nanog is excluded from the nucleus). 多能性幹細胞(PSC)を、DMSO、ZVAD(100nM)、BRQ(10μM)又はBRQ及びZVAD(BRQ:10μM、ZVAD:100nM)の存在下にて2日間培養し、Nanog及びOct4を免疫標識した結果を示す、蛍光顕微鏡写真である。図中、「ESC」及び「iPSC」は、マウスES細胞及びマウスiPS細胞を観察した結果を示す。図中の数値は、全細胞における各核PSCマーカー陽性細胞数の割合を示す。上段の8写真は、Oct4(緑色)を免疫染色した結果を示し、中段の8写真は、Nanog(赤色)を免疫染色した結果を示し、下段の8写真は、前記Oct4を免疫染色した結果及びNanogを免疫染色した結果に、DAPIによって細胞核(青色)を対比染色した結果を重ね合わせた結果を示す。スケールバーは50μmを示す。Pluripotent stem cells (PSC) were cultured for 2 days in the presence of DMSO, ZVAD (100 nM), BRQ (10 μM) or BRQ and ZVAD (BRQ:10 μM, ZVAD:100 nM), and immunolabeled with Nanog and Oct4. It is a fluorescence micrograph which shows a result. In the figure, “ESC” and “iPSC” indicate the results of observing mouse ES cells and mouse iPS cells. The numerical values in the figure show the ratio of the number of cells positive for each nuclear PSC marker in all cells. The upper 8 photographs show the results of immunostaining Oct4 (green), the middle 8 photographs show the results of immunostaining Nanog (red), and the lower 8 photographs show the results of immunostaining Oct4 and The result of superimposing the result of counter-staining the cell nucleus (blue) with DAPI on the result of immunostaining Nanog is shown. The scale bar shows 50 μm. 多能性幹細胞(PSC)を、DMSO、BRQ(10μM)又はBRQ及びLMB(BRQ:10μM、LMB:0.15nM)の存在下にて2日間培養し、Nanog(赤色)及びOct4(緑色)を免疫標識した結果を示す、蛍光顕微鏡写真である。図中、「ESC」及び「iPSC」は、マウスES細胞及びマウスiPS細胞を各々観察した結果を示す。図中の数値は、全細胞における各核PSCマーカー陽性細胞数の割合を示す。上段の6写真は、Oct4(緑色)を免疫染色した結果を示し、中段の6写真は、Nanog(赤色)を免疫染色した結果を示し、下段の6写真は、前記Oct4を免疫染色した結果及びNanogを免疫染色した結果に、DAPIによって細胞核(青色)を対比染色した結果を重ね合わせた結果を示す。スケールバーは100μmを示す。Pluripotent stem cells (PSC) were cultured in the presence of DMSO, BRQ (10 μM) or BRQ and LMB (BRQ:10 μM, LMB: 0.15 nM) for 2 days, and Nanog (red) and Oct4 (green) were cultured. It is a fluorescence micrograph which shows the result of immunolabeling. In the figure, "ESC" and "iPSC" indicate the results of observing mouse ES cells and mouse iPS cells, respectively. The numerical values in the figure show the ratio of the number of cells positive for each nuclear PSC marker in all cells. The upper 6 photographs show the results of immunostaining Oct4 (green), the middle 6 photographs show the results of immunostaining Nanog (red), and the lower 6 photographs show the results of immunostaining Oct4 and The results of immunostaining Nanog with the results of counterstaining the cell nuclei (blue) with DAPI are shown. The scale bar shows 100 μm. DMSO又はBRQにより前処理した多能性幹細胞から形成されたテラトーマの代表的な写真を示す。図中、「ESC」及び「iPSC」は、マウスES細胞及びマウスiPS細胞各々から形成されたテラトーマを観察した結果を示す。スケールバーは1cmを示す。Representative photographs of teratomas formed from pluripotent stem cells pretreated with DMSO or BRQ are shown. In the figure, “ESC” and “iPSC” indicate the results of observing teratomas formed from mouse ES cells and mouse iPS cells, respectively. Scale bar indicates 1 cm. DMSO又はBRQにより前処理した多能性幹細胞から形成された腫瘍の体積を示すグラフである。図中、「ESC」及び「iPSC」は、マウスES細胞及びマウスiPS細胞各々から形成された腫瘍の体積を示す。エラーバーは±SDを示す。統計学的有意性はt検定によって判定した。**P<0.01,***P<0.001。FIG. 6 is a graph showing the volume of tumors formed from pluripotent stem cells pretreated with DMSO or BRQ. In the figure, “ESC” and “iPSC” indicate the volume of tumor formed from mouse ES cells and mouse iPS cells, respectively. Error bars indicate ±SD. Statistical significance was determined by t-test. ***P<0.01, ***P<0.001. DMSO又はBRQにより処理したマウスにおいて、多能性幹細胞から形成されたテラトーマの代表的な写真を示す。図中、「ESC」及び「iPSC」は、マウスES細胞及びマウスiPS細胞各々から形成されたテラトーマを観察した結果を示す。スケールバーは1cmを示す。Representative photographs of teratomas formed from pluripotent stem cells in mice treated with DMSO or BRQ are shown. In the figure, “ESC” and “iPSC” indicate the results of observing teratomas formed from mouse ES cells and mouse iPS cells, respectively. Scale bar indicates 1 cm. DMSO又はBRQにより処理したマウスにおいて、多能性幹細胞から形成されたテラトーマの腫瘍体積を示すグラフである。図中、「ESC」及び「iPSC」は、マウスES細胞及びマウスiPS細胞各々から形成された腫瘍体積を示す。エラーバーは±SDを示す。統計学的有意性はt検定によって判定した。**P<0.01,***P<0.001。FIG. 6 is a graph showing the tumor volume of teratomas formed from pluripotent stem cells in mice treated with DMSO or BRQ. In the figure, “ESC” and “iPSC” indicate tumor volumes formed from mouse ES cells and mouse iPS cells, respectively. Error bars indicate ±SD. Statistical significance was determined by t-test. ***P<0.01, ***P<0.001. DMSO又はBRQにより処理したマウスにおいて、多能性幹細胞から形成されたテラトーマにおけるKi67陽性細胞の割合示すグラフである。図中、「ESC」及び「iPSC」は、マウスES細胞及びマウスiPS細胞各々から形成されたテラトーマにおける前記割合を示す。エラーバーは±SDを示す。統計学的有意性はt検定によって判定した。**P<0.01,***P<0.001。6 is a graph showing the proportion of Ki67-positive cells in teratomas formed from pluripotent stem cells in mice treated with DMSO or BRQ. In the figure, "ESC" and "iPSC" indicate the above-mentioned proportions in teratoma formed from mouse ES cells and mouse iPS cells, respectively. Error bars indicate ±SD. Statistical significance was determined by t-test. ***P<0.01, ***P<0.001. Oct4(赤色)、Nanog(緑色)、AFP(緑色)、βIIIチューブリン(赤色)及びSMA(緑色)を対象とする免疫染色を施したテラトーマについての代表的な画像を示す。図中、「ESC」及び「iPSC」は、マウスES細胞及びマウスiPS細胞各々から形成されたテラトーマを観察した結果を示す。核はDAPI(青色)にて対比染色によって検出した。スケールバーは100μmを示す。Representative images of teratoma immunostained for Oct4 (red), Nanog (green), AFP (green), βIII tubulin (red) and SMA (green) are shown. In the figure, “ESC” and “iPSC” indicate the results of observing teratomas formed from mouse ES cells and mouse iPS cells, respectively. Nuclei were detected by counterstaining with DAPI (blue). The scale bar shows 100 μm.
 後述の実施例に示すとおり、本発明者は、ジヒドロオロト酸デヒドロゲナーゼ(DHODH)の酵素活性を阻害し、ピリミジン合成を阻害することによって、多能性幹細胞に細胞傷害をもたらし、当該細胞を除去できることを明らかにした。また、その一方で、多能性幹細胞を分化誘導することによって得られえる体性幹細胞や体細胞等の分化細胞においては、DHODHを阻害しても、有意な細胞傷害は生じないことも見出した。 As shown in Examples described below, the present inventors have shown that by inhibiting the enzymatic activity of dihydroorotate dehydrogenase (DHODH) and inhibiting pyrimidine synthesis, pluripotent stem cells can be cytotoxicized and removed. Revealed. On the other hand, it was also found that in somatic stem cells obtained by inducing differentiation of pluripotent stem cells or differentiated cells such as somatic cells, even if DHODH is inhibited, significant cytotoxicity does not occur. ..
 したがって、本発明は、DHODH阻害剤を用いる、多能性幹細胞から分化誘導した細胞群に残存する未分化の多能性幹細胞を除去するための組成物及び方法を、提供する。 Accordingly, the present invention provides a composition and a method for removing undifferentiated pluripotent stem cells remaining in a cell population differentiated from pluripotent stem cells using a DHODH inhibitor.
 本発明において、「ジヒドロオロト酸デヒドロゲナーゼ(DHODH)」は、ピリミジンのde novo合成経路において第4反応であるジヒドロオロト酸からオロト酸への酸化を触媒する酵素を意味し、例えば、ジヒドロオロト酸デヒドロゲナーゼ(フマル酸)(EC番号:1.3.98.1)、ジヒドロオロト酸デヒドロゲナーゼ(NAD+)(EC番号:1.3.1.14)、ジヒドロオロト酸デヒドロゲナーゼ(NADP+)(EC番号:1.3.1.15)、ジヒドロオロト酸デヒドロゲナーゼ(キノン)(EC番号:1.3.5.2)が挙げられる。 In the present invention, “dihydroorotic acid dehydrogenase (DHODH)” means an enzyme that catalyzes the oxidation of dihydroorotic acid to orotic acid, which is the fourth reaction in the de novo synthetic pathway of pyrimidine, and includes, for example, dihydroorotic acid dehydrogenase (fumaric acid). ) (EC number: 1.3.98.1), dihydroorotic acid dehydrogenase (NAD+) (EC number: 1.3.1.14), dihydroorotic acid dehydrogenase (NADP+) (EC number: 1.3.1.15). ), and dihydroorotic acid dehydrogenase (quinone) (EC number: 1.3.5.2).
 「ジヒドロオロト酸デヒドロゲナーゼ阻害剤〈DHODH阻害剤)」は、前記触媒反応を阻害する活性を有する化合物を意味する。なお、本発明における「阻害」には活性等の完全な阻害のみならず部分的な阻害(抑制)も含まれる。 “Dihydroorotic acid dehydrogenase inhibitor (DHODH inhibitor)” means a compound having an activity of inhibiting the catalytic reaction. The “inhibition” in the present invention includes not only complete inhibition of activity and the like but also partial inhibition (suppression).
 本発明にかかる「DHODH阻害剤」としては、後述の実施例に示すような、ブレキナー(brequinar、BRQ、6-フルオロ-2-(2’-フルオロ-1,1’-ビフェニル-4-イル)-3-メチル-4-キノリン-カルボン酸ナトリウム塩)、レフルノミド(leflunomide、5-メチル-N-[4-(トリフルオロメチル)フェニル]-イソオキサゾール-4-カルボキサミド)、テリフルノミド(teriflunomide、(2Z)-2-シアノ-3-ヒドロキシ-N-[4-(トリフルオロメチル)フェニル]ブタ-2-エンアミド)、ビドフルジムス(vidofludimus、2-(3-フルオロ-3’-メトキシビフェニル-4-イルカルバモイル)-シクロペンタ-1-エンカルボン酸、ASLAN003(2-(3,5-ジフルオロ-3’メトキシビフェニル-4-イルアミノ)ニコチン酸)等が挙げられる。さらに、例えば、J Med Chem.、2013 Apr 25;56(8):3148-67に開示されている化合物15~110、Eur J Med Chem.2012 Mar;49:102-9に開示されている化合物7a~7n、Biochemistry.2008 Aug 26;47(34):8929-36に開示されている化合物 コード3~8、国際公開第2008/077639号に開示されているアミノニコチン及びイソニコチン酸誘導体、Neuro Oncol.2019 Sep 10.pii:noz170.doi:10.1093/neuonc/noz170.に開示されている化合物 10580(後述のIC50:9nM)が挙げられる。 Examples of the “DHODH inhibitor” according to the present invention include brequinar (BRQ, 6-fluoro-2-(2′-fluoro-1,1′-biphenyl-4-yl), as shown in Examples below. -3-methyl-4-quinoline-carboxylic acid sodium salt), leflunomide (leflunomide, 5-methyl-N-[4-(trifluoromethyl)phenyl]-isoxazole-4-carboxamide), teriflunomide, (2Z )-2-Cyano-3-hydroxy-N-[4-(trifluoromethyl)phenyl]but-2-enamide), Vidofludimus, 2-(3-Fluoro-3′-methoxybiphenyl-4-ylcarbamoyl) )-Cyclopent-1-enecarboxylic acid, ASLAN003 (2-(3,5-difluoro-3′methoxybiphenyl-4-ylamino)nicotinic acid), etc. Further, for example, J Med Chem., 2013 Apr 25 56(8):3148-67, compounds 15-110, Eur J Med Chem. 2012 Mar; 49:102-9, compounds 7a-7n, Biochemistry. 2008 Aug 26;47(; 34): 8929-36, Codes 3-8, aminonicotine and isonicotinic acid derivatives disclosed in WO 2008/077639, Neuro Oncol. 2019 Sep 10. pii: noz170.doi: Compound 10580 (IC 50 : 9 nM described later) disclosed in 10.1093/neuonc/noz170.
 本発明にかかる「DHODH阻害剤」として、好ましくはヒト由来DHODHに対する50%阻害濃度(IC50)が100nM以下である化合物が好ましく、IC50が50nM以下である化合物がより好ましく、IC50が20nM以下である化合物がさらに好ましい。かかる化合物としては、例えば、以下の化合物が挙げられる(各化合物の構造及びヒト由来DHODHに対するIC50を以下に示す)。 The “DHODH inhibitor” according to the present invention is preferably a compound having a 50% inhibitory concentration (IC 50 ) for human-derived DHODH of 100 nM or less, more preferably a compound having an IC 50 of 50 nM or less, and an IC 50 of 20 nM. The following compounds are more preferable. Examples of such compounds include the following compounds (the structure of each compound and the IC 50 for human-derived DHODH are shown below).
Figure JPOXMLDOC01-appb-C000001
Figure JPOXMLDOC01-appb-C000001
Figure JPOXMLDOC01-appb-C000002
Figure JPOXMLDOC01-appb-C000002
Figure JPOXMLDOC01-appb-C000003
Figure JPOXMLDOC01-appb-C000003
Figure JPOXMLDOC01-appb-C000004
Figure JPOXMLDOC01-appb-C000004
Figure JPOXMLDOC01-appb-C000005
Figure JPOXMLDOC01-appb-C000005
Figure JPOXMLDOC01-appb-C000006
Figure JPOXMLDOC01-appb-C000006
Figure JPOXMLDOC01-appb-C000007
Figure JPOXMLDOC01-appb-C000007
 DHODHの酵素活性は、当業者であれば、例えば、ジクロロインドフェノール(DCIP)を用いる色素還元アッセイにより求めることができる。吸収極大を600nmに持つDCIPは、DHODHによる基質の酸化及び電子受容体の還元に伴い、還元され無色となる。そのため、当該波長における吸光度の減少を指標として、DHODHの活性を求めることができる。また、DHODHに対するIC50は、例えば、ヒト由来のDHODH、基質(ジヒドロオロト酸)、電子受容体(例えば、CoQ)、及び緩衝液を含む酵素反応液に、様々な濃度の供試化合物(DHODH阻害剤)を添加し、前記DCIPアッセイを行なうことにより求めることができる(国際公開第2008/077639号、Biochem J.1998 Dec 1;336(Pt2):299-303 参照のほど)。 The enzyme activity of DHODH can be determined by those skilled in the art by, for example, a dye reduction assay using dichloroindophenol (DCIP). DCIP having an absorption maximum at 600 nm is reduced and becomes colorless due to the oxidation of the substrate by DHODH and the reduction of the electron acceptor. Therefore, the activity of DHODH can be determined using the decrease in the absorbance at the wavelength as an index. In addition, the IC 50 for DHODH is, for example, that DOHDH derived from human, a substrate (dihydroorotic acid), an electron acceptor (for example, CoQ), and an enzyme reaction solution containing a buffer have various concentrations of a test compound (DHODH inhibition). Agent) and perform the DCIP assay (see WO 2008/077639, Biochem J. 1998 Dec 1;336(Pt2):299-303).
 また、本発明にかかる「DHODH阻害剤」には、その阻害活性を有する限り、薬理学上許容される塩、水和物又は溶媒和物も含まれる。このような薬理学上許容される塩としては、特に制限はなく、当該薬剤の各構造等に応じて適宜選択することができ、例えば、酸付加塩(塩酸塩、硫酸塩、臭化水素塩、硝酸塩、硫酸水素酸塩、リン酸塩、酢酸塩等)、塩基付加塩(ナトリウム塩、カリウム塩、亜鉛塩、カルシウム塩等)が挙げられる。また、水和物又は溶媒和物としては、特に制限はなく、例えば、DHODH阻害剤又はその塩1分子に対し、0.1~10分子の水又は溶媒が付加したものが挙げられる。 The “DHODH inhibitor” according to the present invention also includes a pharmacologically acceptable salt, hydrate or solvate as long as it has the inhibitory activity. Such a pharmacologically acceptable salt is not particularly limited and can be appropriately selected according to each structure of the drug, and examples thereof include acid addition salts (hydrochloride, sulfate, hydrobromide). , Nitrates, hydrogensulfates, phosphates, acetates, etc.) and base addition salts (sodium salts, potassium salts, zinc salts, calcium salts, etc.). The hydrate or solvate is not particularly limited, and examples thereof include those in which 0.1 to 10 molecules of water or solvent are added to 1 molecule of DHODH inhibitor or its salt.
 さらに、本発明にかかる「DHODH阻害剤」には、その阻害活性を有する限り、互変異性体、幾何異性体、不斉炭素に基づく光学異性体、立体異性体等の総ての異性体及び異性体混合物が含まれる。さらにまた、DHODH阻害剤が生体内で酸化、還元、加水分解、アミノ化、脱アミノ化、水酸化、リン酸化、脱水酸化、アルキル化、脱アルキル化、抱合等の代謝を受けてなおその阻害活性を示す化合物をも包含し、また本発明は生体内で酸化、還元、加水分解等の代謝を受けてDHODH阻害剤を生成する化合物をも包含する。 Furthermore, the “DHODH inhibitor” according to the present invention includes all isomers such as tautomers, geometric isomers, optical isomers based on asymmetric carbons, stereoisomers and the like, as long as it has the inhibitory activity. Includes isomer mixtures. Furthermore, DHODH inhibitors undergo metabolism in vivo such as oxidation, reduction, hydrolysis, amination, deamination, hydroxylation, phosphorylation, dehydroxylation, alkylation, dealkylation, conjugation, etc., and still inhibit the metabolism. The present invention also includes compounds that exhibit activity, and the present invention also includes compounds that are metabolized in vivo by oxidation, reduction, hydrolysis and the like to produce DHODH inhibitors.
 なお、DHODH阻害剤に関しては、上述のとおり、多くの化合物が開発され市販もされているため、購入することによって入手することができる。また市販されていなくとも、それら化合物の製造方法についても多々報告がなされている。そのため、当業者であれば、当該製造方法に沿って適宜調製することもできる。 Regarding the DHODH inhibitor, many compounds have been developed and are on the market as described above, so they can be obtained by purchasing. Many reports have been made on methods for producing these compounds even if they are not commercially available. Therefore, a person skilled in the art can also appropriately prepare according to the manufacturing method.
 本発明において「多能性幹細胞」とは、分化万能性及び自己複製能を備えている細胞であればよく、例えば、胚性幹細胞(ES細胞)、胚性腫瘍細胞(EC細胞)、エピブラスト幹細胞(EpiS細胞)、胚性生殖細胞(EG細胞)、多能性生殖細胞(mGS細胞)、MUSE細胞(Kuroda Y.ら、Proc.Natl.Acad.Sci.U.S.A.、2010年、107巻、19号、8639~8643ページ 参照)等の生体から採取され得る細胞が挙げられる。さらに、「多能性幹細胞」には、人工多能性幹細胞(iPS細胞)のように、生体から採取された体細胞から人工的に分化多能性等を持たせるよう誘導された細胞も含まれる。また、これら細胞の由来としては特に制限はなく、ヒト及び非ヒト動物(例えば、マウス及びラット等のげっ歯類、ウシ、ウマ、ブタ、ヒツジ、サル、イヌ、並びにネコ等の哺乳類、ニワトリ等の鳥類)が挙げられる。さらに、後述の再生医療等を目的とする場合には、免疫拒絶反応を抑えるという観点から後述の分化細胞を移植する対象に由来する多能性幹細胞であることが望ましい。 In the present invention, the “pluripotent stem cell” may be any cell having pluripotency and self-renewal ability, and examples thereof include embryonic stem cells (ES cells), embryonal tumor cells (EC cells), and epiblasts. Stem cells (EpiS cells), embryonic germ cells (EG cells), pluripotent germ cells (mGS cells), MUSE cells (Kuroda Y. et al., Proc. Natl. Acad. Sci. USA, 2010) , Vol. 107, No. 19, pp. 8639-8643). Furthermore, "pluripotent stem cells" also include cells derived from somatic cells collected from a living body so as to have artificial pluripotency, such as induced pluripotent stem cells (iPS cells). Be done. The origin of these cells is not particularly limited, and humans and non-human animals (for example, rodents such as mice and rats, cows, horses, pigs, sheep, monkeys, dogs, mammals such as cats, chickens, etc.) Birds). Furthermore, in the case of aiming at the regenerative medicine described below, pluripotent stem cells derived from the target to which the differentiated cells described below are transplanted are desirable from the viewpoint of suppressing immune rejection.
 本発明において「未分化の多能性幹細胞」としては、分化しておらず、分化万能性を維持している前述の多能性幹細胞の他、多能性幹細胞が腫瘍化した細胞(例えば、テラトーマ、テトラカルシノーマ)も含まれる。 In the present invention, "undifferentiated pluripotent stem cells" include, in addition to the pluripotent stem cells that have not been differentiated and maintain pluripotency, cells in which pluripotent stem cells have turned into tumors (for example, Teratoma, tetracarcinoma) are also included.
 本発明において「多能性幹細胞から分化誘導した細胞群」とは、上述の多能性幹細胞に由来する任意の分化細胞を含む細胞の集団を意味する。かかる細胞群は、単種の分化細胞を含むものであってもよく、複数種の分化細胞を含むものであってもよい。さらに、その形態としては、それら細胞によって構成される組織、器官であってもよい。 In the present invention, the “cell group differentiated from pluripotent stem cells” means a population of cells including any differentiated cells derived from the above pluripotent stem cells. Such a cell group may include a single type of differentiated cell or may include a plurality of types of differentiated cells. Further, the form may be a tissue or an organ composed of these cells.
 本発明において「分化細胞」には、多能性を完全に喪失した細胞(最終分化細胞、例えば、体細胞、生殖細胞)のみならず、多能性を部分的に喪失した細胞(例えば、前駆細胞、体性幹細胞)も含まれる。「体性幹細胞」は、成体幹細胞、組織幹細胞とも称される細胞であり、例えば、神経幹細胞、衛星細胞、造血幹細胞(骨髄幹細胞)、間葉系幹細胞、腸管幹細胞、毛包幹細胞、乳腺幹細胞、内皮幹細胞、嗅粘膜幹細胞、神経冠幹細胞、精巣細胞が挙げられる。 In the present invention, "differentiated cells" include not only cells that have completely lost pluripotency (terminally differentiated cells, such as somatic cells and germ cells) but also cells that have partially lost pluripotency (eg, progenitor cells). Cells, somatic stem cells) are also included. "Somatic stem cells" are cells also called adult stem cells and tissue stem cells, and include, for example, neural stem cells, satellite cells, hematopoietic stem cells (bone marrow stem cells), mesenchymal stem cells, intestinal stem cells, hair follicle stem cells, mammary stem cells, Examples include endothelial stem cells, olfactory mucosa stem cells, neural crest stem cells, and testis cells.
 「分化細胞」への多能性幹細胞からの分化誘導は、通常、各分化細胞が発生する胚葉(外胚葉、中胚葉、内胚葉)への分化誘導を介して行なわれ、当業者であれば、各胚葉系細胞への分化誘導に適した低分子化合物、タンパク質等(分化誘導因子)を用いた公知の方法を、適宜選択して行なうことができる。例えば、神経細胞等の外胚葉系細胞へは、BMP阻害剤、TGFβ及びアクチビン阻害剤の存在下にて培養することによって分化誘導することができる(Nat Biotechnol.2009 Mar;27(3):275-80 参照)。軟骨細胞等の中胚葉系の細胞へは、例えば、多能性幹細胞を、Wnt及びアクチビンの存在下にて培養した後、BMP4の存在下にて培養することにより、分化誘導することができる(Development.2008 Sep;135(17):2969-79 参照)。また例えば、膵臓細胞、腸細胞等の内胚葉系細胞は、多能性幹細胞をアクチビンの存在下にて培養した後、BMP4の存在下にて培養することによって、分化誘導することができる(例えば、Diabetes 2010 Sep;59(9):2094-2101、Nature. 2011 Feb 3;470(7332):105-9 参照)。 Induction of differentiation of pluripotent stem cells into "differentiated cells" is usually carried out through induction of differentiation into germ layers (ectodermal, mesoderm, endoderm) in which each differentiated cell develops. A known method using a low-molecular compound, a protein or the like (differentiation inducing factor) suitable for inducing differentiation into each germ layer cell can be appropriately selected and performed. For example, ectodermal cells such as nerve cells can be induced to differentiate by culturing in the presence of a BMP inhibitor, TGFβ and an activin inhibitor (Nat Biotechnol. 2009 Mar; 27(3):275. -80). Differentiation can be induced into mesodermal cells such as chondrocytes by culturing pluripotent stem cells in the presence of Wnt and activin, and then culturing them in the presence of BMP4 ( Development.2008 Sep;135(17):2969-79). Further, for example, endodermal cells such as pancreatic cells and intestinal cells can be induced to differentiate by culturing pluripotent stem cells in the presence of activin and then culturing them in the presence of BMP4 (for example, , Diabetes 2010 Sep; 59(9):2094-2101, Nature. 2011 Feb 3; 470(7332): 105-9).
 また、本発明において、上述の細胞群に残存する未分化の多能性幹細胞の「除去」は、当該細胞の完全な除去のみならず、当該細胞の数又は前記細胞群における割合の低減を意味する。また、当該除去は、in vitroで行なわれるものであってもよく、in vivo又はex vivoで行なわれるものであってもよい。 Further, in the present invention, “removal” of undifferentiated pluripotent stem cells remaining in the above-mentioned cell group means not only complete removal of the cells but also reduction of the number of the cells or the ratio in the cell group. To do. In addition, the removal may be performed in vitro, or may be performed in vivo or ex vivo.
 (多能性幹細胞を除去するための組成物)
 本発明の実施形態として、DHODH阻害剤を有効成分として含有する、多能性幹細胞から分化誘導した細胞群に残存する未分化の多能性幹細胞を除去するための組成物が挙げられる。
(Composition for removing pluripotent stem cells)
Embodiments of the present invention include a composition containing a DHODH inhibitor as an active ingredient for removing undifferentiated pluripotent stem cells remaining in a cell group differentiated from pluripotent stem cells.
 本発明の組成物は、上述のDHODH阻害剤の他、生理学的に許容される担体を含むものであってもよい。生理学的に許容される担体としては、例えば、生理的な等張液(生理食塩水、培地、ブドウ糖やその他の補助薬(D-ソルビトール、D-マンニトール、塩化ナトリウム等)を含む等張液等)、賦形剤、防腐剤、安定剤(ヒト血清アルブミン、ポリエチレングリコール等)、結合剤、溶解補助剤、非イオン性界面活性剤、緩衝剤(リン酸塩緩衝液、酢酸ナトリウム緩衝液等)、保存剤、酸化防止剤が挙げられる。 The composition of the present invention may contain a physiologically acceptable carrier in addition to the above-mentioned DHODH inhibitor. Examples of physiologically acceptable carriers include physiological isotonic solutions (physiological saline, culture medium, glucose and other isotonic solutions containing D-sorbitol, D-mannitol, sodium chloride, etc.). ), excipients, preservatives, stabilizers (human serum albumin, polyethylene glycol, etc.), binders, solubilizing agents, nonionic surfactants, buffers (phosphate buffer, sodium acetate buffer, etc.) , Preservatives and antioxidants.
 本発明の組成物は、研究目的又は医療目的(例えば、分化細胞、特に腫瘍化リスクの低減された分化細胞の製造)に用いられる試薬の形態であり得る。また、後述のとおり、前記分化細胞を移植した対象に投与される、医薬組成物の形態でもあり得る。 The composition of the present invention may be in the form of a reagent used for research purposes or medical purposes (for example, production of differentiated cells, particularly differentiated cells with reduced tumorigenicity risk). Further, as described below, it may be in the form of a pharmaceutical composition to be administered to a subject into which the differentiated cells have been transplanted.
 また、本発明は、分化細胞、特に腫瘍化リスクの低減された分化細胞を製造するためのキットをも提供する。当該キットにおいては、前記組成物の他、多能性幹細胞、上述の分化誘導因子、分化誘導用培地、培養器等の、多能性幹細胞から所望の細胞への分化誘導に必要な材料を含めることができる。さらに、所望の細胞に分化誘導できたことを確認するための、当該細胞特異的マーカー分子を検出するための試薬(例えば、当該マーカー分子に対して特異的な抗体)も、本発明のキットに含めることもできる。また、本発明の組成物の使用方法、分化誘導方法等を示した説明書も本発明のキットに含まれる。 The present invention also provides a kit for producing a differentiated cell, particularly a differentiated cell with reduced tumorigenicity risk. In the kit, in addition to the composition, pluripotent stem cells, the above-mentioned differentiation-inducing factor, differentiation-inducing medium, incubator, and other materials necessary for inducing differentiation of pluripotent stem cells into desired cells are included. be able to. Furthermore, a reagent for detecting the cell-specific marker molecule (for example, an antibody specific to the marker molecule) for confirming that differentiation into desired cells can be induced is also included in the kit of the present invention. It can also be included. In addition, the kit of the present invention also includes instructions indicating the method of using the composition of the present invention, the method of inducing differentiation and the like.
 (多能性幹細胞を除去するための方法)
 本発明の実施形態としてはまた、多能性幹細胞から分化誘導した細胞群とジヒドロオロト酸デヒドロゲナーゼ阻害剤とを接触させる工程を含む、前記細胞群から残存する未分化の多能性幹細胞を除去する方法が挙げられる。
(Method for removing pluripotent stem cells)
The embodiment of the present invention also includes a step of contacting a group of cells differentiated from pluripotent stem cells with a dihydroorotic acid dehydrogenase inhibitor, a method of removing undifferentiated pluripotent stem cells remaining from the group of cells Are listed.
 上述のDHODH阻害剤と細胞群との「接触」については特に制限はなく、例えば、当該細胞群を維持するための培地にDHODH阻害剤を添加することによって行なうことができる。その際の添加濃度としては、特に制限はなく、当業者であれば、対象とする多能性幹細胞及び細胞群の種類、並びに用いるDHODH阻害剤の種類等によって適宜調整することができるが、通常0.1~1000μMであり、好ましくは1~100μM、さらに好ましくは10~50μMである。 There is no particular limitation on the “contact” between the above-mentioned DHODH inhibitor and the cell group, and for example, it can be performed by adding the DHODH inhibitor to the medium for maintaining the cell group. The concentration to be added at that time is not particularly limited and can be appropriately adjusted by those skilled in the art depending on the types of the pluripotent stem cells and cell groups of interest, the type of DHODH inhibitor to be used, etc. It is 0.1 to 1000 μM, preferably 1 to 100 μM, and more preferably 10 to 50 μM.
 なお、このように未分化の多能性幹細胞を除去することにより、再生医療等に有用な、腫瘍化リスクの低減された分化細胞を得ることができる。したがって、本発明の方法は、「多能性幹細胞から分化誘導した細胞群とジヒドロオロト酸デヒドロゲナーゼ阻害剤とを接触させる工程を含む、腫瘍化リスクの低減された分化細胞を製造する方法」とも言い換えることができる。 By removing undifferentiated pluripotent stem cells in this manner, differentiated cells with a reduced risk of tumorigenesis, which are useful for regenerative medicine and the like, can be obtained. Therefore, the method of the present invention can be rephrased as "a method for producing differentiated cells with reduced tumorigenicity risk, which comprises the step of contacting a cell group differentiated from pluripotent stem cells with a dihydroorotate dehydrogenase inhibitor" You can
 また、再生医療等のため、前記分化細胞を対象(ヒト又は非ヒト動物)に移植した場合には、DHODH阻害剤を当該対象に投与することによって、上述のDHODH阻害剤と細胞群との「接触」を行なうことができる。投与形態としては特に制限はなく、静脈内投与、動脈内投与、腹腔内投与、皮下投与、皮内投与、気道内投与、直腸投与及び筋肉内投与、輸液による投与、局所投与、経口投与が挙げられ、移植する細胞群の種類、移植部位等に応じて適宜選択される。また、投与量は、対象の種類、年齢、体重、症状及び健康状態、投与形態、対象とする多能性幹細胞及び細胞群の種類、並びに用いるDHODH阻害剤の種類等によって適宜調整されるが、1回あたりの投与量は、通常、0.001~1000mg/kg体重であり、好ましくは、0.1~100mg/kg体重であり、より好ましくは、1~50mg/kg体重である。また、1日あたりの投与回数としても、特に制限はなく、前記のような様々な要因を考慮して、適宜調整される。さらに、対象への投与は継続して行なわれてもよい(例えば、1週間毎に1回投与)。 Further, when the differentiated cells are transplanted into a subject (human or non-human animal) for regenerative medicine or the like, the DHODH inhibitor and the cell group can be treated by administering the DHODH inhibitor to the subject. Contact can be made. The dosage form is not particularly limited, and examples thereof include intravenous administration, intraarterial administration, intraperitoneal administration, subcutaneous administration, intradermal administration, respiratory tract administration, rectal administration and intramuscular administration, administration by infusion, local administration, and oral administration. It is appropriately selected depending on the type of cell group to be transplanted, the site of transplantation and the like. The dose is appropriately adjusted depending on the type of subject, age, weight, symptoms and health condition, administration form, types of target pluripotent stem cells and cell groups, types of DHODH inhibitors used, and the like. The dose per administration is usually 0.001 to 1000 mg/kg body weight, preferably 0.1 to 100 mg/kg body weight, and more preferably 1 to 50 mg/kg body weight. The number of administrations per day is also not particularly limited, and may be adjusted appropriately in consideration of various factors as described above. Further, the subject may be continuously administered (eg, once a week).
 また、未分化の多能性幹細胞を除去するための、DHODH阻害剤と細胞群との接触時間としては、当該除去が十分に行なわれる時間であればよく、当業者であれば適宜調整することができるが、通常2~7日間であり、好ましくは2~5日間であり、より好ましくは2~3日間である。 Further, the contact time between the DHODH inhibitor and the cell population for removing the undifferentiated pluripotent stem cells may be any time sufficient for the removal, and those skilled in the art can appropriately adjust the time. However, it is usually 2 to 7 days, preferably 2 to 5 days, and more preferably 2 to 3 days.
 以下、実施例に基づいて本発明をより具体的に説明するが、本発明は以下の実施例に限定されるものではない。また、本実施例は、以下の材料及び方法を用いて行なった。 Hereinafter, the present invention will be described more specifically based on examples, but the present invention is not limited to the following examples. In addition, this example was performed using the following materials and methods.
 (動物・化学試薬)
 マウスを用いた全ての実験は、北海道大学動物実験委員会が承認したプロトコールに従って行った。マウスは株式会社ホクドーから購入したものを用いた。特に断りがない限り、化学薬品及び増殖因子は、各々Invitrogen社及びPeproTech社から購入したものを用いた。
(Animal/Chemical reagents)
All experiments using mice were performed according to the protocol approved by the Hokkaido University Animal Experiment Committee. The mouse used was purchased from Hokudo Co., Ltd. Unless otherwise specified, chemicals and growth factors used were those purchased from Invitrogen and PeproTech, respectively.
 (多能性幹細胞)
 マウスES細胞は、升井伸治氏(当該細胞を譲り受け時、京都大学iPS研究所に在籍)より譲り受けた細胞株(E14tg2a)を用いた。マウスiPS細胞は、理化学研究所バイオリソース研究センターから購入した細胞株(iPS-Hep-FB/Ng/gfp-103c-1)を用いた。
(Pluripotent stem cells)
As the mouse ES cells, a cell line (E14tg2a) given by Mr. Shinji Masui (at the time of receiving the cells, enrolled in Kyoto University iPS Laboratory) was used. As the mouse iPS cells, a cell line (iPS-Hep-FB/Ng/gfp-103c-1) purchased from RIKEN BioResource Research Center was used.
 これら多能性幹細胞は、SIGMA社のウェブサイトで公開している方法にて培養した(https://www.sigmaaldrich.com/life-science/stem-cell-biology/stem-cell-protocols.html 参照)。また培養には、以下の組成からなる培養液(ESC培養液)を用いた。
1mM ピルビン酸ナトリウム(Gibco社製,カタログ番号:11360-070)、1x非必須アミノ酸(Gibco社製,カタログ番号11140-050)、100μM 2-メルカプトエタノール(SIGMA社製,CAS番号:60-24-2)、1000units/mL 白血病阻害因子(LIF、ESGRO(登録商標),Gibco社製,カタログ番号:ESG1107)、15% ノックアウト血清代替物(KSR、Thermo Fisher Scientific社製,カタログ番号:10828028)、及び1% ウシ胎仔血清(FBS、Hyclone社製)を含む、グラスゴー最小必須培地(GMEM培地、Sigma社製,製品番号:G5154-500ML)。さらに、これら細胞を維持培養するために、100mm培養ディッシュ(Falcon社製)の表面を、ウシ表皮由来のゼラチン(Sigma社製,製品番号:G9391)にてコートし、37℃にて2時間以上置いたものを用いた。
These pluripotent stem cells were cultured by the method disclosed on the SIGMA website (https://www.sigmaaldrich.com/life-science/stem-cell-biology/system-cell-protocols.html). reference). For the culture, a culture medium (ESC culture medium) having the following composition was used.
1 mM sodium pyruvate (manufactured by Gibco, catalog number: 11360-070), 1x non-essential amino acid (manufactured by Gibco, catalog number 11140-050), 100 μM 2-mercaptoethanol (manufactured by SIGMA, CAS number: 60-24-) 2), 1000 units/mL leukemia inhibitory factor (LIF, ESGRO (registered trademark), manufactured by Gibco, catalog number: ESG1107), 15% knockout serum substitute (KSR, manufactured by Thermo Fisher Scientific, catalog number: 10828028), and Glasgow minimum essential medium (GMEM medium, Sigma, product number: G5154-500ML) containing 1% fetal bovine serum (FBS, Hyclone). Furthermore, in order to maintain and culture these cells, the surface of a 100 mm culture dish (manufactured by Falcon) was coated with gelatin derived from bovine epidermis (manufactured by Sigma, product number: G9391) and kept at 37° C. for 2 hours or more. The placed one was used.
 ヒト胚性癌(EC)細胞)(NT2)は、American Tissue Culture Collectionから入手し(CRL-1973)、10%FBS含有DMEM(ダルベッコ改変イーグル培地)にて培養した。 Human embryonal carcinoma (EC) cells (NT2) were obtained from the American Tissue Culture Collection (CRL-1973) and cultured in DMEM (Dulbecco's modified Eagle medium) containing 10% FBS.
 (分化細胞)
 マウス神経幹細胞(mNSC)は、Johe KKら、Genes Dev.1996年、10巻、24号、3129~3140ページに記載のとおり、マウス(胎仔14.5日目)の終脳から調製し、NSC培養液[bFGF(10ng/ml)、EGF(10ng/ml)、ヘパリン(5μM)、GlutaMAX(Gibco社製)、ペニシリン、及びストレプトマイシンを含むDMEM/F12培地(Sigma社製)]にて培養した。
(Differentiated cells)
Mouse neural stem cells (mNSCs) are described in Johe KK et al., Genes Dev. 1996, Vol. 10, No. 24, pp. 3129-3140, prepared from the telencephalon of mice (fetus 14.5 days), and cultured in NSC medium [bFGF (10 ng/ml), EGF (10 ng/ml). ), heparin (5 μM), GlutaMAX (manufactured by Gibco), penicillin, and streptomycin in DMEM/F12 medium (manufactured by Sigma)].
 マウス骨髄由来ストローマ細胞(PA6)は、理研バイオリソースセンターから入手し(RBRC-RCB1127)、10%FBS含有αMEM(イーグル最小必須培地 α改変型)にて培養した。 Mouse bone marrow-derived stromal cells (PA6) were obtained from Riken BioResource Center (RBRC-RCB1127) and cultured in αMEM containing 10% FBS (Eagle minimum essential medium α modified type).
 マウス筋芽細胞(C2C12)は、理研バイオリソースセンターから入手し(RBRC-RCB0987)、10%FBS含有DMEMにて培養した。 Mouse myoblasts (C2C12) were obtained from Riken BioResource Center (RBRC-RCB0987) and cultured in DMEM containing 10% FBS.
 アストロサイトは、mNSCを10%FBS含有DMEM存在下で3日間培養することにより調製、その後、同じ培地で維持した。 Astrocytes were prepared by culturing mNSC in the presence of DMEM containing 10% FBS for 3 days, and then maintained in the same medium.
 (MTTアッセイ法)
 96ウェルプレートの各ウェルに、1000個の各細胞を播種し、DMSOのみを終濃度0.1%になるよう添加した培地(200μL/ウェル)にて、又は各ジヒドロオロト酸デヒドロゲナーゼ(DHODH)阻害剤を添加した培地(200μL/ウェル)にて、3日間培養した。なお、培地は、上述の各細胞に適した培養液を用いた。
(MTT assay method)
1000 cells were seeded in each well of a 96-well plate and DMSO alone was added to a medium (200 μL/well) to a final concentration of 0.1%, or each dihydroorotic acid dehydrogenase (DHODH) inhibitor The cells were cultured for 3 days in a medium (200 μL/well) supplemented with. The culture medium used was a culture medium suitable for each of the cells described above.
 DHODH阻害剤として、Brequnar(BRQ,Cayman Chemical,CAS番号:96187-53-0),Leflunomide(Cayman Chemical,CAS番号:75706-12-6),Teriflunomide(Cayman Chemical,CAS番号:163451-81-8)又はVidofludimus(Cayman,CAS番号:717824-30-1)を、培地に添加した。各DHODH阻害剤の培地への添加濃度は、200μMからの2倍希釈系列とした。 As a DHODH inhibitor, Brequnar (BRQ, Cayman Chemical, CAS number: 96187-53-0), Leflunomide (Cayman Chemical, CAS number: 75706-12-6), Teriflunomide (Cayman45:Chamical 81ChemicalChemical number: 81:Chamman: 45-81-Chemical number: 45-81-Chemical number: 16-Chemical-Chemical number) ) Or Vidofludimus (Cayman, CAS number: 717824-30-1) was added to the medium. The concentration of each DHODH inhibitor added to the medium was a 2-fold dilution series starting from 200 μM.
 また、バックグランドとして、培地のみを入れたウェルを用意し、さらにコントロールとして、TritonX-100(終濃度0.2%)を含む培地を細胞に添加したウェルも用意した。 As a background, a well containing only the medium was prepared, and as a control, a well containing a medium containing Triton X-100 (final concentration 0.2%) was added to the cells.
 そして、各ウェルにMTT(5mg/ml、Wako社製)5μLを添加し、細胞を37℃で2~3時間インキュベートした。次いで、培地を100μLのDMSOに置換して細胞を溶解し、それら細胞溶解液の波長570nmにおける吸光度を、マイクロプレートリーダー(Bio-Rad社製、Benchmarkモデル)にて測定した。また、得られた吸光度に基づき、細胞生存率は、以下の式によって算出した。
細胞生存率(%)=(DHODH阻害剤添加ウェルにおける吸光度-バックグランドにおける吸光度)/(コントロールにおける吸光度-バックグランドにおける吸光度)×100。
Then, 5 μL of MTT (5 mg/ml, manufactured by Wako) was added to each well, and the cells were incubated at 37° C. for 2 to 3 hours. Next, the medium was replaced with 100 μL of DMSO to lyse the cells, and the absorbance of the cell lysate at a wavelength of 570 nm was measured with a microplate reader (Bio-Rad, Benchmark model). The cell viability was calculated by the following formula based on the obtained absorbance.
Cell viability (%)=(absorbance in DHODH inhibitor-added well-absorbance in background)/(absorbance in control-absorbance in background)×100.
 (ヌクレオシド中和実験)
 ES細胞又はiPS細胞を、BRQ(10μM)の存在下、又はBRQ(10μM)及び様々な濃度のリボヌクレオシド(~500μM、全てSigma社製)の存在下にて、3日間インキュベートし、上述のMTTアッセイにより細胞生存率を分析した。
(Nucleoside neutralization experiment)
ES cells or iPS cells were incubated for 3 days in the presence of BRQ (10 μM) or in the presence of BRQ (10 μM) and various concentrations of ribonucleosides (up to 500 μM, all manufactured by Sigma), and the MTT described above was added. Cell viability was analyzed by assay.
 (免疫染色)
 細胞を、Kondoら、Genes Dev.2004年、18巻、23号、2963~72ページに記載の方法にて、固定し、免疫染色を施した。具体的には先ず、免疫染色に際して、細胞を、コーティングした8ウェルチャンバースライド(ガラス)上に播種し、10μM BRQの存在下で24時間培養した。なお、mNSCの培養には、PDL+フィブロネクチンコートを施したものを用いた(Johe KKら、Genes Dev.1996年、10巻、24号、3129~3140ページ 参照)。ES細胞及びiPS細胞の培養にはゼラチンコートを施したものを用いた。
(Immunostaining)
Cells were cultured in Kondo et al., Genes Dev. It was fixed and immunostained by the method described in 2004, Vol. 18, No. 23, pp. 2963-72. Specifically, first, for immunostaining, cells were seeded on a coated 8-well chamber slide (glass) and cultured in the presence of 10 μM BRQ for 24 hours. For mNSC culture, PDL+fibronectin-coated cells were used (see Joe KK et al., Genes Dev. 1996, 10, 24, 3129-3140). Gelatin-coated cells were used for the culture of ES cells and iPS cells.
 そして、免疫染色においては先ず、PBSにて細胞を1回洗浄した後、4%パラフォルムアルデヒドを加えて室温で10分間インキュベートすることにより固定した。0.3%TritonX100含有PBSで置換して室温で5分間インキュベートした後、0.3%TritonX100及び10%FCS含有PBS(ブロッキング溶液)に置換して室温で30分間インキュベートした。ブロッキング溶液で希釈した1次抗体溶液に置換し、室温で2時間インキュベートした。PBSで3回洗浄した後、ブロッキング溶液で希釈した2次抗体及びDAPI溶液を加えて、室温で2時間インキュベートした。PBSで3回洗浄した後、退色防止剤(DAKO社製、コード番号:S3023)で封入し、顕微鏡観察を行った。なお、DAPI溶液(1μg/mL、Dojindo社製、製品コード:D523)による対比染色によって、細胞核を可視化した。また、蛍光画像は、AxioImager M1顕微鏡(Carl Zeiss社製)にて取得した。 Then, in the immunostaining, first, the cells were washed once with PBS, added with 4% paraformaldehyde, and incubated at room temperature for 10 minutes for fixation. After replacing with PBS containing 0.3% Triton X100 and incubating at room temperature for 5 minutes, it was replaced with PBS containing 0.3% Triton X100 and 10% FCS (blocking solution) and incubated at room temperature for 30 minutes. The solution was replaced with a primary antibody solution diluted with a blocking solution, and incubated at room temperature for 2 hours. After washing three times with PBS, the secondary antibody diluted with the blocking solution and the DAPI solution were added, and the mixture was incubated at room temperature for 2 hours. After washing three times with PBS, the cells were encapsulated with an anti-fading agent (manufactured by DAKO, code number: S3023) and observed under a microscope. The cell nuclei were visualized by counterstaining with a DAPI solution (1 μg/mL, manufactured by Dojindo, product code: D523). The fluorescence image was acquired with an AxioImager M1 microscope (Carl Zeiss).
 上記培養細胞に対する免疫染色において、抗原の検出には以下の抗体を用いた。
一次抗体:
抗Nestinマウスモノクローナル抗体(BD社製,クローン Rat401、1:200に希釈して使用)、
抗Nanogウサギポリクローナル抗体(Wako pure chemical社製、コード番号:018-27521、1:200に希釈して使用)、
抗Sox2ウサギポリクローナル抗体(StemCell Technology社製、1:500に希釈して使用)、
抗Oct4マウスモノクローナル抗体(Cell Signaling Tech社製、1:200に希釈して使用)、
抗GFPラットモノクローナル抗体(ナカライテスク社製、1:500に希釈して使用)、
抗Ki67ウサギモノクローナル抗体(Thermo Fisher Scientific社製、1:100に希釈して使用)、
抗活性型Caspase3ウサギポリクローナル抗体(StemCell Technology社製、1:1000に希釈して使用)。
二次抗体:
Alexa488標識抗マウスIgGロバポリクローナル抗体(Thermo Fisher Scientific社製、カタログ番号:R37120、1:500に希釈して使用)、
Alexa488標識抗ウサギIgGロバポリクローナル抗体(Thermo Fisher Scientific社製、カタログ番号:A-11008、1:500に希釈して使用)、
Alexa594標識抗ウサギIgGロバポリクローナル抗体(Thermo Fisher Scientific社製、カタログ番号:R37119、1:500に希釈して使用)、
Alexa594標識抗マウスIgGロバポリクローナル抗体(Thermo Fisher Scientific社製、カタログ番号:R37115、1:500に希釈して使用)、
Alexa488標識抗ラットIgGヤギ抗体(Jackson ImmunoResearch社製、1:500に希釈して使用)。
In the immunostaining of the above-mentioned cultured cells, the following antibodies were used to detect the antigen.
Primary antibody:
Anti-Nestin mouse monoclonal antibody (BD, clone Rat401, diluted 1:200 for use),
Anti-Nanog rabbit polyclonal antibody (manufactured by Wako pure chemical, code number: 018-27521, used by diluting 1:200),
Anti-Sox2 rabbit polyclonal antibody (manufactured by StemCell Technology, diluted 1:500 for use),
Anti-Oct4 mouse monoclonal antibody (Cell Signaling Tech, diluted 1:200 for use),
Anti-GFP rat monoclonal antibody (manufactured by Nacalai Tesque, Inc., diluted 1:500),
Anti-Ki67 rabbit monoclonal antibody (Thermo Fisher Scientific, manufactured at a dilution of 1:100),
Anti-active Caspase3 rabbit polyclonal antibody (manufactured by StemCell Technology, diluted 1:1000 and used).
Secondary antibody:
Alexa488-labeled anti-mouse IgG donkey polyclonal antibody (Thermo Fisher Scientific, catalog number: R37120, diluted 1:500 for use),
Alexa488-labeled anti-rabbit IgG donkey polyclonal antibody (Thermo Fisher Scientific, catalog number: A-11008, diluted 1:500 for use),
Alexa 594-labeled anti-rabbit IgG donkey polyclonal antibody (Thermo Fisher Scientific, catalog number: R37119, diluted 1:500 for use),
Alexa 594-labeled anti-mouse IgG donkey polyclonal antibody (Thermo Fisher Scientific, catalog number: R37115, diluted 1:500 for use),
Alexa488 labeled anti-rat IgG goat antibody (manufactured by Jackson ImmunoResearch, diluted 1:500 and used).
 腫瘍については、厚さ10μmの切片を調製し、Kondo T,及びRaff M.、EMBO J.2000;19(9):1998-2007、並びにTakanaga H,ら、Stem Cells.2009;27(1):165-174.に記載の方法にて免疫染色を行った。当該免疫染色には、以下の抗体を用いた。
一次抗体:
抗Oct4マウスモノクローナル抗体(Cell Signaling Tech社製、1:200に希釈して使用)、
抗Nanogウサギポリクローナル抗体(Wako Pure Chemical社製、1:200に希釈して使用)、
抗Ki67ウサギモノクローナル抗体(Thermo Fisher Scientific社製、1:100に希釈して使用)、
抗βIIIチューブリンマウスモノクローナル抗体(Sigma社製、1:400に希釈して使用)、
抗平滑筋アクチンウサギポリクローナル抗体(Proteintech社製、SMA、1:200に希釈して使用)、
抗αフェトプロテインウサギポリクローナル抗体(Proteintech社製、AFP、1:100に希釈して使用)。
二次抗体:
Alexa594標識抗マウスIgGロバポリクローナル抗体(Thermo Fisher Scientific社製、カタログ番号:R37115、1:500に希釈して使用)、
Alexa488標識抗ウサギIgGヤギ抗体(Jackson ImmunoResearch社製、1:500に希釈して使用)。
For tumors, 10 μm thick sections were prepared and examined by Kondo T, and Raff M. et al. EMBO J.; 2000;19(9):1998-2007, and Takanaga H, et al., Stem Cells. 2009;27(1):165-174. Immunostaining was performed by the method described in 1. The following antibodies were used for the immunostaining.
Primary antibody:
Anti-Oct4 mouse monoclonal antibody (Cell Signaling Tech, diluted 1:200 for use),
Anti-Nanog rabbit polyclonal antibody (manufactured by Wako Pure Chemical, diluted 1:200 for use),
Anti-Ki67 rabbit monoclonal antibody (Thermo Fisher Scientific, manufactured at a dilution of 1:100),
Anti-βIII tubulin mouse monoclonal antibody (Sigma, diluted 1:400 for use),
Anti-smooth muscle actin rabbit polyclonal antibody (manufactured by Proteintech, SMA, diluted 1:200 for use),
Anti-alpha fetoprotein rabbit polyclonal antibody (Proteintech, AFP, diluted 1:100 for use).
Secondary antibody:
Alexa 594-labeled anti-mouse IgG donkey polyclonal antibody (Thermo Fisher Scientific, catalog number: R37115, diluted 1:500 for use),
Alexa488-labeled anti-rabbit IgG goat antibody (Jackson ImmunoResearch, 1:500 dilution).
 (混合培養)
 マウスNSCと、マウスES細胞又はマウスiPS細胞とを、各々2000個ずつ播種し、1μM又は10μMのBRQ存在下、ESC培養液にて3日間培養した。そして、上述の免疫染色にて分析した。
(Mixed culture)
2000 mouse NSCs and mouse ES cells or mouse iPS cells were seeded, and cultured in the ESC culture medium for 3 days in the presence of 1 μM or 10 μM BRQ. And it analyzed by the above-mentioned immunostaining.
 (定量RT-PCR)
 マウスES細胞及びマウスiPS細胞を、上述のウシ表皮由来のゼラチンにてコーティングした100mm培養ディッシュ及びESC培養液にて、BRQ存在下又は非存在下培養した後、これら細胞における、Sox2、Oct4及びNanogの遺伝子発現量を、以下に示す方法にて測定した。
(Quantitative RT-PCR)
Mouse ES cells and mouse iPS cells were cultured in the above-described 100 mm culture dish coated with bovine epidermis-derived gelatin and ESC culture medium in the presence or absence of BRQ, and then Sox2, Oct4, and Nanog in these cells were cultured. The gene expression level was measured by the following method.
 先ず、細胞からRNeasyキット(Qiagen社製)を用いてトータルRNAを調製し、トランスクリプターファーストストランドcDNA合成キット(Roche Applied Science社製)を用いてcDNAを合成した。 First, total RNA was prepared from cells using an RNeasy kit (manufactured by Qiagen), and cDNA was synthesized using a transcript first-strand cDNA synthesis kit (manufactured by Roche Applied Science).
 そして、当該cDNAを鋳型とし、サンダーバード サイバー qPCRミックス(TOYOBO社製)及びステップワンプラス(Thermo Fisher Scientific社製)を用い、リアルタイムPCRを行なった。反応条件は、95℃15秒、60℃30秒を40サイクルとし、以下に示すオリゴヌクレオチドプライマーを使用し、各標的遺伝子を増幅した。
Sox2遺伝子:
フォワードプライマー 5’-TGAAGAAGGATAAGTACACGCT-3’(配列番号:1)、
リバースプライマー 5’-TCCTGCATCATGCTGTAGCTG-3’(配列番号:2)、
oct4遺伝子:
フォワードプライマー 5’-CTGAAGCAGAAGAGGATCACC-3’(配列番号:3)、
リバースプライマー 5’-CCGCAGCTTACACATGTTCTT-3’(配列番号:4)、
nanog遺伝子:
フォワードプライマー 5’-CTGATTCTTCTACCAGTCCCAA-3’(配列番号:5)、
リバースプライマー 5’-AGAGTTCTTGCATCTGCTGGA-3’(配列番号:6)、
18SリボゾームRNA遺伝子:
フォワードプライマー 5’-CGGACAGGATTGACAGATTG-3’(配列番号:7)、
リバースプライマー 5’-CAAATCGCTCCACCAACTAA-3’(配列番号:8)。
なお、このようにして検出された各標的遺伝子の発現量は、18SリボゾームRNAの発量をもとに相対定量法(delta delta Ct法)により正規化した。
Then, using the cDNA as a template, real-time PCR was performed using Thunderbird Cyber qPCR mix (manufactured by TOYOBO) and Step One Plus (manufactured by Thermo Fisher Scientific). The reaction conditions were 95° C. for 15 seconds and 60° C. for 30 seconds for 40 cycles, and each target gene was amplified using the oligonucleotide primers shown below.
Sox2 gene:
Forward primer 5'-TGAAGAAGGAATAAGTACACCGCT-3' (SEQ ID NO: 1),
Reverse primer 5'-TCCTGCATCATGCTGTAGCTG-3' (SEQ ID NO: 2),
oct4 gene:
Forward primer 5′-CTGAAGCAGAAGAGGATCACC-3′ (SEQ ID NO: 3),
Reverse primer 5'-CCGCAGCTTACACATGTTCTT-3' (SEQ ID NO: 4),
nanog gene:
Forward primer 5'-CTGATTCTTCTACCAGTCCCCAA-3' (SEQ ID NO: 5),
Reverse primer 5'-AGAGTTCTTGCATCTGCTGGA-3' (SEQ ID NO: 6),
18S ribosomal RNA gene:
Forward primer 5′-CGGACAGGATTGACAGATTG-3′ (SEQ ID NO: 7),
Reverse primer 5'-CAAATCGCTCCACCAACTAA-3' (SEQ ID NO: 8).
The expression level of each target gene thus detected was normalized by the relative quantification method (delta delta Ct method) based on the amount of 18S ribosomal RNA generated.
 (ベクター)
 ベクターは、Nishide K,ら、PLoS ONE.2009;4(8):e6869.及びTakanaga H,ら、Stem Cells.2009;27(1):165-174.の記載に沿って構築した。
(vector)
Vectors are based on Nishide K, et al., PLoS ONE. 2009; 4(8):e6869. And Takanaga H, et al., Stem Cells. 2009;27(1):165-174. It was constructed according to the description.
 具体的には先ず、マウスDHODHの全長cDNAは、KOD Plus-Ver.2ポリメラーゼ(東洋紡株式会社製)を用い、その使用説明書に沿って、マウスNSC由来cDNAから増幅した。当該増幅には、5’プライマー(5’-AGAATTCAATGGGGGAACA-3’ (配列番号:9))及び3’プライマー(5’-TTGGATTCTCTGCGGTC-3’(配列番号:10))を用いた。増幅したcDNAを、p3xFLAG CMV10ベクターに挿入し、p3xFLAG CMV10-mDHODHを調製した。 Specifically, first, the full-length cDNA of mouse DHODH is KOD Plus-Ver. 2 polymerase (manufactured by Toyobo Co., Ltd.) was used and amplified from mouse NSC-derived cDNA according to the instruction manual. A 5'primer (5'-AGAATTCAATGGGGGAACA-3' (SEQ ID NO: 9)) and a 3'primer (5'-TTGGATTCTCTGCCGGTC-3' (SEQ ID NO: 10)) were used for the amplification. The amplified cDNA was inserted into the p3xFLAG CMV10 vector to prepare p3xFLAG CMV10-mDHODH.
 また、マウスDHODHをノックダウンするために、InvivoGenのsiRN Wizardソフトウェア(http://www.sirnawizard.com/)を用い、当該遺伝子を標的とする3つのショートヘアピン(sh)配列を選択した。これらsh配列をpsiRNA-h7SKhygro G1発現ベクター(InvivoGen社製)に各々挿入し、psiRNA-h7SKhygro-mDHODHsh1-3を調製した。これらベクターによるノックダウン効率はウェスタンブロッティングにより分析した(図14 参照)。その結果、高い効果を示したDHODH sh1及びsh3をノックダウン実験に用いた。 To knockdown mouse DHODH, we used invivoGen's siRN Wizard software (http://www.sirnawizard.com/) to select three short hairpin (sh) sequences targeting the gene. These sh sequences were inserted into psiRNA-h7SKhygro G1 expression vector (manufactured by InvivoGen) to prepare psiRNA-h7SKhygro-mDHODHsh1-3. The knockdown efficiency of these vectors was analyzed by Western blotting (see Fig. 14). As a result, DHODH sh1 and sh3, which showed a high effect, were used in the knockdown experiment.
 マウスDHODHを対象とするsh1及びのsh3の標的配列は、それぞれ5’-GGCTAGCTGTCtCTCTCT-3’(配列番号:11)及び5’-GGAAGCTGTGTCTCTCtA-3’(配列番号:12)である。コントロールとして用いたsh(egfp)の標的配列は5’-GCAAGCTGACCCGTGTTCA-3’(配列番号:13)である。 The target sequences of sh1 and sh3 targeting mouse DHODH are 5'-GGCTAGCTGTCtCTCTCT-3' (SEQ ID NO: 11) and 5'-GGAAGCTGTGTCTCTCA-3' (SEQ ID NO: 12), respectively. The target sequence of sh(egfp) used as a control is 5'-GCAAGCTGACCCGTGTTTCA-3' (SEQ ID NO: 13).
 クローンニングしたcDNAの塩基配列は、BigDyeターミネーターキットバージョン3.1(Applied Biosystems社製)及びABIシーケンサーモデル3130xl(Applied Biosystems社製)を用いて確認した。 The nucleotide sequence of the cloned cDNA was confirmed using BigDye terminator kit version 3.1 (manufactured by Applied Biosystems) and ABI sequencer model 3130xl (manufactured by Applied Biosystems).
 また、ベクターは、リポフェクタミン3000(Thermo Fisher Scientific社製)を用い、その使用説明書の記載に沿って、細胞に導入した。 Also, Lipofectamine 3000 (Thermo Fisher Scientific Co., Ltd.) was used as a vector, and the vector was introduced into cells according to the instruction manual.
 (テラトーマ形成)
 多能性幹細胞を、BRQの存在下又は非存在下で2日間培養した。生存細胞(1x10)を、50μlのマトリゲル(BD Biosciences社製)に懸濁し、10%ペントバルビタールで麻酔した、5~8週齢のNOD/SCIDマウスの臀部に皮下注射した。そして、注射してから4週後に、マウスを犠牲死させ、Tsukamoto Y,ら、Stem Cells.2016;34(8):2016-2025.に記載の方法に沿って、腫瘍を摘出して撮影し、それらのサイズを測定した。また得られた測定値に基づき、腫瘍の体積は、以下の式によって算出した。
腫瘍の体積(cm)=長径×短径×短径×1/2。
(Teratoma formation)
Pluripotent stem cells were cultured for 2 days in the presence or absence of BRQ. Viable cells (1×10 6 ) were suspended in 50 μl of Matrigel (manufactured by BD Biosciences) and anesthetized with 10% pentobarbital, and subcutaneously injected into the buttocks of 5-8 week old NOD/SCID mice. Then, 4 weeks after the injection, the mice were sacrificed, and Tsukamoto Y, et al., Stem Cells. 2016;34(8):2016-2025. Tumors were excised, photographed, and their sizes were measured according to the method described in 1. Further, the volume of the tumor was calculated by the following formula based on the obtained measured value.
Tumor volume (cm 3) = long diameter × short diameter × short diameter × 1/2.
 また、BRQのテラトーマ形成抑制活性を調べるために、皮下腫瘍を有するマウスの腹腔内に、200μlの生理食塩水、又は25mg/kgブレキナーナトリウム塩(BRQ、TOCRIS社製)PBS溶液 200μlを、毎日投与した。腹腔内投与の5日後に、マウスを犠牲死させ、Tsukamoto Y,ら、Stem Cells.2016;34(8):2016-2025.に記載の方法に沿って、腫瘍を摘出して撮影し、病理学的分析に供した。 Further, in order to examine the teratoma formation inhibitory activity of BRQ, 200 μl of physiological saline or 25 μg/kg of Brekiner sodium salt (BRQ, TOCRIS) PBS solution 200 μl was intraperitoneally injected into the abdominal cavity of mice with subcutaneous tumor every day. Was administered. Five days after the intraperitoneal administration, the mice were sacrificed, and Tsukamoto Y, et al., Stem Cells. 2016;34(8):2016-2025. The tumor was excised, photographed and subjected to pathological analysis according to the method described in 1.
 (ウェスタンブロッティング)
 ウェスタンブロッティングは、Takanaga H,ら、Stem Cells.2009;27(1):165-174.に記載の方法に沿って行なった。また、ウェスタンブロッティング及び免疫沈降には、一次抗体として、抗FLAG M2マウス抗体(SIGMA社製、10μ/ml)及び抗GAPDH抗体(Proteintech社製、1:5000に希釈して使用)を用い、二次抗体として、西洋ワサビペルオキシダーゼ標識抗マウスIgG抗体(Santa Cruz社製、1:5000に希釈して使用)を用いた。
(Western blotting)
Western blotting is described by Takanaga H, et al., Stem Cells. 2009;27(1):165-174. Was carried out according to the method described in. For Western blotting and immunoprecipitation, an anti-FLAG M2 mouse antibody (manufactured by SIGMA, 10 μ/ml) and an anti-GAPDH antibody (manufactured by Proteintech, diluted 1:5000 and used) were used as primary antibodies. As a secondary antibody, a horseradish peroxidase-labeled anti-mouse IgG antibody (manufactured by Santa Cruz, diluted 1:5000 for use) was used.
 (統計解析)
 上記にて得られた細胞生存率及び遺伝子発現量等における2群間の比較は、student t-検定にて解析した。また、カプラン-マイヤー曲線は、無調整のイベント発生迄の時間(unadjusted time-to-event variables)を推定するために用いた。P値が0.05未満(両側検定)である場合、有意であると判断した。
(Statistical analysis)
The comparison between the two groups in the cell viability and the gene expression level obtained above was analyzed by the student t-test. The Kaplan-Meier curve was also used to estimate the unadjusted time-to-event variables. A P value of less than 0.05 (two-sided test) was considered significant.
 (実施例1) 多能性幹細胞に対するDHODH阻害剤の細胞傷害活性についての検討
 マウスES細胞及びマウスiPS細胞を、DHODH阻害剤(BRQ、Leflunomide、Teriflunomide又はVidefludimus)の存在下で各々3日間培養し、生存率をMTTアッセイにより検討した。
(Example 1) Examination on cytotoxic activity of DHODH inhibitor against pluripotent stem cells Mouse ES cells and mouse iPS cells were cultured for 3 days in the presence of DHODH inhibitors (BRQ, Leflunomide, Teriflunomide or Videfludimus), respectively. , The survival rate was examined by the MTT assay.
 その結果、図1及び2に示すとおり、試験した4種のDHODH阻害剤は全て、両多能性幹細胞に対する細胞傷害活性を有していることが明らかになった。特に、BRQに関しては、低濃度(10μM~)でも、多能性幹細胞に対する有意な細胞傷害活性が認められた。なお、BRQ、Leflunomide、Teriflunomide及びVidofludimusのヒト由来のDHODHに対するIC50は各々、6~20nM、98μM、1μM及び134nMである。そのため、DHODH阻害剤間における多能性幹細胞に対する細胞傷害活性の差は、阻害活性の差に起因しているように見受けられる。 As a result, as shown in FIGS. 1 and 2, it was revealed that all of the four types of DHODH inhibitors tested had cytotoxic activity against pluripotent stem cells. In particular, regarding BRQ, significant cytotoxic activity against pluripotent stem cells was observed even at low concentrations (10 μM-). The IC 50 of BRQ, Leflunomide, Teriflunomide, and Vidofludimus for human-derived DHODH are 6 to 20 nM, 98 μM, 1 μM, and 134 nM, respectively. Therefore, the difference in cytotoxic activity against pluripotent stem cells between DHODH inhibitors seems to be due to the difference in inhibitory activity.
 (実施例2) 体性幹細胞等に対するDHODH阻害剤の細胞傷害活性についての検討
 マウス正常神経幹細胞(mNSC)、5%ウシ胎仔血清存在下で培養したmNSC(mNSC+5%FCS)、マウスアストロサイトを、10μM BRQ存在下で3日間培養し、生存率をMTTアッセイにより検討した。その結果、図3に示すとおり、神経幹細胞及び神経細胞(分化細胞)はBRQに対して低感受性であることが明らかになった。
(Example 2) Examination on cytotoxic activity of DHODH inhibitor on somatic stem cells, etc. Mouse normal neural stem cells (mNSC), mNSCs cultured in the presence of 5% fetal calf serum (mNSC+5% FCS), mouse astrocytes, After culturing for 3 days in the presence of 10 μM BRQ, the survival rate was examined by MTT assay. As a result, as shown in FIG. 3, it was revealed that neural stem cells and neural cells (differentiated cells) have low sensitivity to BRQ.
 また、他の多能性幹細胞(NT2:ヒト胚性癌(EC)細胞)及び他の体性幹細胞(PA6:マウス骨髄由来ストローマ細胞、C2C12:マウス筋芽細胞)についても、10μM BRQ存在下で3日間培養し、生存率をMTTアッセイにより検討した。その結果、図4に示すとおり、BRQは多能性幹細胞(ES細胞、iPS細胞及びEC細胞)では顕著な細胞傷害活性を示した。一方、ES細胞及びiPS細胞の生存率と、PA6細胞、C2C12細胞、神経幹細胞及びアストロサイトのそれとの間に統計学的有意差が認められた(全てp<0.01)。すなわち、BRQの体性幹細胞及び体細胞に対する有意な細胞傷害活性は認められなかった。 In addition, other pluripotent stem cells (NT2: human embryonal carcinoma (EC) cells) and other somatic stem cells (PA6: mouse bone marrow-derived stromal cells, C2C12: mouse myoblasts) in the presence of 10 μM BRQ After culturing for 3 days, the survival rate was examined by MTT assay. As a result, as shown in FIG. 4, BRQ showed remarkable cytotoxic activity in pluripotent stem cells (ES cells, iPS cells and EC cells). On the other hand, a statistically significant difference was observed between the survival rate of ES cells and iPS cells and that of PA6 cells, C2C12 cells, neural stem cells and astrocytes (all p<0.01). That is, no significant cytotoxic activity of BRQ on somatic stem cells and somatic cells was observed.
 (実施例3) 多能性幹細胞に対するDHODH阻害剤の細胞特異的傷害活性についての検証
 多能性幹細胞(マウスES細胞又はマウスiPS細胞)と、該細胞から分化誘導して得られる細胞と見立てた体性幹細胞(マウス神経幹細胞)とを、様々な濃度のBRQを含むES細胞培地にて3日間混合培養し、NSCマーカーであるNestin及び多能性幹細胞マーカーであるNanogに対する免疫染色を行った。その結果、図5及び6に示すとおり、上述の図1~4に示した結果同様に、10μMのBRQ存在下でES細胞とiPS細胞は消失したが、神経幹細胞への明らかな細胞傷害は観られなかった。
(Example 3) Verification of cell-specific damaging activity of DHODH inhibitor on pluripotent stem cells Pluripotent stem cells (mouse ES cells or mouse iPS cells) and cells obtained by inducing differentiation of the cells were regarded as the cells. Somatic stem cells (mouse neural stem cells) were mixed and cultured in an ES cell medium containing various concentrations of BRQ for 3 days, and immunostained for NSC Nestin and pluripotent stem cell marker Nanog. As a result, as shown in FIGS. 5 and 6, like the results shown in FIGS. 1 to 4 above, ES cells and iPS cells disappeared in the presence of 10 μM BRQ, but no apparent cytotoxicity to neural stem cells was observed. I couldn't do it.
 (実施例4) DHODH阻害剤の細胞傷害に対する、ヌクレオシド中和活性についての検証
 ピリミジン合成は、新生(de novoの合成)経路と再利用(サルベージ)経路とによって制御されている。DHODHは、当該合成経路において律速となる4番目の化学反応を触媒する、de novo合成のキー因子である。そこで、DHODH阻害剤による多能性幹細胞の除去効果が、サルベージ経路の出発材料であるウリジン等のヌクレオシドを添加することにより、当該効果を解消できるかどうかを調べた。より具体的には、BRQの存在下、ヌクレオシド(アデノシン、グアノシン、シチジン又はウリジン)を培地に添加することによって、当該阻害剤による多能性幹細胞の細胞傷害活性を中和し得るかを検証した。その結果、図7及び8に示すとおり、BRQの細胞傷害活性に対し、ピリミジンヌクレオシド(ウリジン、シチジン)は中和活性を示し、特にウリジンは強い中和活性を示すことが明らかになった。
Example 4 Verification of Nucleoside Neutralizing Activity Against Cytotoxicity of DHODH Inhibitors Pyrimidine synthesis is regulated by nascent (de novo synthesis) and salvage (salvage) pathways. DHODH is a key factor in de novo synthesis that catalyzes the fourth rate-limiting chemical reaction in the synthetic pathway. Therefore, it was investigated whether the effect of removing pluripotent stem cells by the DHODH inhibitor can be eliminated by adding a nucleoside such as uridine which is a starting material of the salvage pathway. More specifically, it was verified by adding a nucleoside (adenosine, guanosine, cytidine or uridine) to the medium in the presence of BRQ that the cytotoxic activity of pluripotent stem cells by the inhibitor can be neutralized. .. As a result, as shown in FIGS. 7 and 8, it was revealed that pyrimidine nucleosides (uridine and cytidine) have a neutralizing activity, and particularly uridine has a strong neutralizing activity against the cytotoxic activity of BRQ.
 また、ピリミジン合成経路の産物であるUMPは、UDP、UTPへと変化する。さらに、UTPを基質としてウリジン二リン酸-N-アセチルグルコサミン(UDP-GlcNac)も生合成される。UDP-GlcNacは、O結合型N-アセチルグルコサミン転移酵素(OGT)の基質となり、細胞内シグナル伝達に広範に関与することが明らかになっている。そこで、DHODH阻害剤による多能性幹細胞の細胞傷害活性が、これらUMPからの生成物によっても中和されるかを検証した。その結果、図9及び10に示すとおり、BRQの細胞傷害活性に対し、UDP及びUDP-GlcNacも中和活性を示すことが明らかになった。 Also, UMP, which is a product of the pyrimidine synthesis pathway, changes to UDP and UTP. Furthermore, uridine diphosphate-N-acetylglucosamine (UDP-GlcNac) is also biosynthesized using UTP as a substrate. It has been revealed that UDP-GlcNac serves as a substrate for O-linked N-acetylglucosaminyltransferase (OGT) and is widely involved in intracellular signal transduction. Therefore, it was examined whether the cytotoxic activity of pluripotent stem cells by the DHODH inhibitor was neutralized by the products from these UMPs. As a result, as shown in FIGS. 9 and 10, it was revealed that UDP and UDP-GlcNac also have a neutralizing activity against the cytotoxic activity of BRQ.
 また、ヌクレオチド合成の中間基質であるヌクレオチド二リン酸(UDP、CDP、ADP、GDP)についても、BRQの細胞傷害活性に対する中和活性を評価した。その結果、上記同様に、UDPにおいて強い中和活性が認められた(図11 参照)。 Also, for the nucleotide diphosphates (UDP, CDP, ADP, GDP), which are intermediate substrates for nucleotide synthesis, the neutralizing activity against the cytotoxic activity of BRQ was evaluated. As a result, similar to the above, a strong neutralizing activity was recognized in UDP (see FIG. 11).
 以上のことから、BRQ等のDHODH阻害剤は、ピリミジン合成経路を阻害することによって、多能性幹細胞に対する細胞傷害活性を奏していることが示唆された。また、その細胞傷害活性は、再利用(サルベージ)経路を活性化することにより抑制できることも示唆された。 From the above, it was suggested that DHODH inhibitors such as BRQ exert cytotoxic activity on pluripotent stem cells by inhibiting the pyrimidine synthesis pathway. It was also suggested that its cytotoxic activity could be suppressed by activating the salvage pathway.
 (実施例5) BRQによる多能性幹細胞における細胞周期停止及び細胞死の誘導についての検証
 多能性幹細胞において、DHODH阻害剤であるBRQがどのような現象を引き起こしているかを明らかにすべく、多能性幹細胞を、BRQの存在下又は非存在下にて2日間培養した後、ブロモ-デオキシウリジン(BrdU)の取り込みアッセイとCasp3免疫染色を行い、それらの細胞増殖性と細胞死を分析した。その結果、図12及び13に示すとおり、BRQは多能性幹細胞の増殖性を低減し、CASP3を強く活性化することが明らかとなった。
Example 5 Verification of Induction of Cell Cycle Arrest and Cell Death in Pluripotent Stem Cells by BRQ To clarify what phenomenon the DHODH inhibitor BRQ causes in pluripotent stem cells, After pluripotent stem cells were cultured in the presence or absence of BRQ for 2 days, bromo-deoxyuridine (BrdU) uptake assay and Casp3 immunostaining were performed to analyze their cell proliferation and cell death. .. As a result, as shown in FIGS. 12 and 13, it was revealed that BRQ reduced the proliferation of pluripotent stem cells and strongly activated CASP3.
 また、BRQ依存性の細胞傷害活性は、DHODH活性阻害に完全に依存しているかどうかを評価すべく、2種類のDHODH特異的shRNA(図14 参照)を用い、DHODHをノックダウンした。その結果、図15~18に示すとおり、多能性幹細胞の増殖性は低減し、CASP3は強く活性化された。このように、BRQ処理細胞同様の現象が、DHODHノックダウンによっても生じたことから、前記BRQ依存性細胞傷害性は、DHODH活性を阻害することによって発揮されたことが示唆された。 Moreover, in order to evaluate whether the BRQ-dependent cytotoxic activity was completely dependent on inhibition of DHODH activity, two types of DHODH-specific shRNA (see FIG. 14) were used to knockdown DHODH. As a result, as shown in FIGS. 15 to 18, proliferation of pluripotent stem cells was reduced and CASP3 was strongly activated. As described above, the phenomenon similar to that of BRQ-treated cells was also caused by DHODH knockdown, suggesting that the BRQ-dependent cytotoxicity was exerted by inhibiting DHODH activity.
 (実施例6) 多能性幹細胞のマーカー分子への、DHODH阻害剤による影響についての検証
 多能性幹細胞においては、自己複製能の促進と未分化状態の維持に関わる3種類の転写因子(Sox2、Oct4、Nanog)が高いレベルにて発現していることが明らかとなっている。そこで、これら多能性幹細胞のマーカー分子へのDHODH阻害剤による影響について検証した。
Example 6 Verification of Effect of DHODH Inhibitor on Marker Molecule of Pluripotent Stem Cell In pluripotent stem cells, three transcription factors (Sox2) involved in promotion of self-renewal ability and maintenance of undifferentiated state. , Oct4, Nanog) are expressed at high levels. Therefore, the effect of the DHODH inhibitor on the marker molecules of these pluripotent stem cells was examined.
 具体的には先ず、BRQ存在下にて24時間培養した、多能性幹細胞(マウスiPS細胞及びマウスES細胞)、並びに体性幹細胞(マウス神経幹細胞)におけるSox2の発現を、蛍光免疫染色にて検出した。その結果、図19及び20に示すとおり、BRQ存在下の24時間培養にて、ES細胞及びiPS細胞では早くも細胞増殖阻害、或いは細胞死が観察された。また、両多能性幹細胞の30%程度においてSox2の発現は認められないようになっていた。一方、体性幹細胞においては同様の変化は認められなかった。 Specifically, first, the expression of Sox2 in pluripotent stem cells (mouse iPS cells and mouse ES cells) and somatic stem cells (mouse neural stem cells) cultured for 24 hours in the presence of BRQ was examined by fluorescent immunostaining. Detected. As a result, as shown in FIGS. 19 and 20, cell growth inhibition or cell death was observed in ES cells and iPS cells as early as in 24-hour culture in the presence of BRQ. In addition, expression of Sox2 was not observed in about 30% of the pluripotent stem cells. On the other hand, similar changes were not observed in somatic stem cells.
 次に、BRQ存在下にて1日又は2日間培養した、多能性幹細胞(マウスiPS細胞及びマウスES細胞)におけるSox2、Oct4及びNanogの遺伝子発現量を、PCRにて測定した。その結果、図21に示すとおり、BRQにて2日間処理した細胞において、多能性幹細胞のマーカー遺伝子の発現量は総じて減少していることが明らかになった。 Next, the gene expression levels of Sox2, Oct4 and Nanog in pluripotent stem cells (mouse iPS cells and mouse ES cells) cultured in the presence of BRQ for 1 day or 2 days were measured by PCR. As a result, as shown in FIG. 21, it was revealed that the expression level of the pluripotent stem cell marker gene was generally decreased in the cells treated with BRQ for 2 days.
 また、BRQ存在下にて2日間培養した、多能性幹細胞(マウスiPS細胞及びマウスES細胞)におけるNanogの発現を、蛍光免疫染色にて検出した。その結果、図22に示すとおり、通常、多能性幹細胞の核に局在しているNanogは、BRQ処理によって細胞質に広がって分布するようになることが明らかになった。 Moreover, the expression of Nanog in pluripotent stem cells (mouse iPS cells and mouse ES cells) cultured for 2 days in the presence of BRQ was detected by fluorescent immunostaining. As a result, as shown in FIG. 22, it was revealed that Nanog, which is normally localized in the nucleus of pluripotent stem cells, becomes spread and distributed in the cytoplasm by BRQ treatment.
 以上の結果から、DHODH阻害剤によって、自己複製能の促進と未分化状態の維持に関わる3種類の転写因子(Sox2、Oct4、Nanog)の発現量が低減し、また細胞内局在が変化することによって、多能性幹細胞に細胞傷害、ひいては細胞死がもたらされることが示唆される。 From the above results, the expression level of three transcription factors (Sox2, Oct4, Nanog) involved in promotion of self-renewal ability and maintenance of undifferentiated state is decreased by the DHODH inhibitor, and intracellular localization is changed. This suggests that pluripotent stem cells undergo cytotoxicity and eventually cell death.
 (実施例7) DHODH阻害剤によって誘導される、CRM1依存的多能性幹細胞マーカー分子の核外輸送
 BRQ若しくはpanカスパーゼ阻害剤であるZ-VADの存在下又は非存在下において、多能性幹細胞を培養した。BRQにて処理した後2日目に、多能性幹細胞(ES細胞及びiPS細胞)におけるOct4陽性細胞及びNanog陽性細胞の数を測定した。
(Example 7) Nuclear export of CRM1-dependent pluripotent stem cell marker molecule induced by DHODH inhibitor Pluripotent stem cells in the presence or absence of BRQ or pan-caspase inhibitor Z-VAD Were cultured. On the second day after treatment with BRQ, the number of Oct4-positive cells and Nanog-positive cells in pluripotent stem cells (ES cells and iPS cells) was measured.
 その結果、図23に示すとおり、BRQにて処理した多能性幹細胞におけるOct4陽性細胞及びNanog陽性細胞の割合は、DMSOにて処理したコントロール多能性幹細胞におけるそれらと比較して、有意に減少した(BRQにて処理したES細胞における、Oct4陽性細胞:2%、Nanog陽性細胞:6%。BRQにて処理したiPS細胞における、Oct4陽性細胞:0%、Nanog陽性細胞:0%。DMSOにて処理したES細胞における、Oct4陽性細胞:85%、Nanog陽性細胞:86%。DMSOにて処理したiPS細胞における、Oct4陽性細胞:94%、Nanog陽性細胞:93%)。 As a result, as shown in FIG. 23, the proportion of Oct4-positive cells and Nanog-positive cells in the BRQ-treated pluripotent stem cells was significantly reduced as compared with those in the DMSO-treated control pluripotent stem cells. (Oct4 positive cells in ES cells treated with BRQ: 2%, Nanog positive cells: 6%. Oct4 positive cells: 0%, Nanog positive cells: 0% in iPS cells treated with BRQ. DMSO Oct4-positive cells: 85%, Nanog-positive cells: 86% in the ES cells treated with, and Oct4-positive cells: 94%, Nanog-positive cells: 93% in the iPS cells treated with DMSO).
 また、Z-VADの添加によって、BRQ依存的細胞死にわずかな遅れを生じたものの、BRQにて処理した多能性幹細胞における前記Nanog陽性細胞及びOct4陽性細胞の割合のの減少を回復させることはなかった(BRQ及びZ-VADにて処理したES細胞における、Oct4陽性細胞:5%、Nanog陽性細胞:2%。BRQ及びZ-VADにて処理したiPS細胞における、Oct4陽性細胞:3%、Nanog陽性細胞:5%)(図23 参照)。 Moreover, although the addition of Z-VAD caused a slight delay in BRQ-dependent cell death, it was possible to restore the decrease in the ratio of Nanog-positive cells and Oct4-positive cells in BRQ-treated pluripotent stem cells. Not present (Oct4 positive cells: 5%, Nanog positive cells: 2% in ES cells treated with BRQ and Z-VAD. Oct4 positive cells: 3% in iPS cells treated with BRQ and Z-VAD, Nanog positive cells: 5%) (see FIG. 23).
 また、BRQ存在下又は非存在下、レプトマイシンB(LMB)を添加した培地にて、多能性幹細胞を培養し、Nanog及びOct4の核局在を分析した。なお、LMBは、Exportin 1としても知られるCRM1に対する特異的阻害剤である。 Also, pluripotent stem cells were cultured in a medium supplemented with leptomycin B (LMB) in the presence or absence of BRQ, and nuclear localization of Nanog and Oct4 was analyzed. LMB is a specific inhibitor of CRM1 also known as Exportin1.
 その結果、図24に示すとおり、LMBは、BRQにて処理した多能性幹細胞におけるNanog及びOct4の核外輸送を完全に抑制した。このことから、BRQによるNanog及びOct4の核からの排除は、CRM1依存的に生じていたことが示唆される。 As a result, as shown in FIG. 24, LMB completely suppressed the nuclear export of Nanog and Oct4 in BRQ-treated pluripotent stem cells. This suggests that the elimination of Nanog and Oct4 from the nucleus by BRQ occurred in a CRM1-dependent manner.
 (実施例8) 多能性幹細胞の腫瘍形成能に対する、DHODH阻害剤の抑制効果についての検証
 DHODH阻害剤であるBRQにて前処理した多能性幹細胞の腫瘍形成能について調べた。具体的には先ず、DMSOのみ又は10μM BRQを添加した培地にて2日間培養した後、生存していた多能性幹細胞をNOD/SCIDマウスの臀部皮下に注入した。そして、移植してから4週間後に、腫瘍を摘出して観察した。
(Example 8) Verification of suppressive effect of DHODH inhibitor on tumorigenicity of pluripotent stem cells The tumorigenicity of pluripotent stem cells pretreated with BRQ which is a DHODH inhibitor was examined. Specifically, first, after culturing in a medium supplemented with DMSO alone or 10 μM BRQ for 2 days, surviving pluripotent stem cells were subcutaneously injected into the buttocks of the NOD/SCID mice. Then, 4 weeks after the transplantation, the tumor was excised and observed.
 その結果、図25及び26に示すとおり、BRQにて前処理した細胞は増殖していなかった。一方、DMSOにて処理した細胞においては、腫瘍形成が認められた。 As a result, as shown in FIGS. 25 and 26, the cells pretreated with BRQ were not proliferating. On the other hand, tumor formation was observed in the cells treated with DMSO.
 また、BRQの抗テラトーマ形成活性を調べるため、先ず、NOD/SCIDマウスの臀部皮下に多能性幹細胞を移植し、100mmを超えるサイズに達する腫瘍を形成させた。そして、3日毎に1回、BRQを腹腔内注射し、5回目の腹腔内注射を施してから3日目に、マウスから腫瘍を摘出した。 In order to examine the anti-teratoma formation activity of BRQ, pluripotent stem cells were first transplanted subcutaneously into the buttocks of NOD/SCID mice to form tumors reaching a size of more than 100 mm 3 . Then, BRQ was intraperitoneally injected once every three days, and a tumor was excised from the mouse on day 3 after the fifth intraperitoneal injection.
 図27に示すとおり、BRQ投与によって腫瘍の増殖は抑制された。なお、図には示さないが、BRQを投与したマウスにおいて目に見える副作用は認められなかった。また図28に示すとおり、DMSO及びBRQにて処理したテラトーマのサイズは各々、ES細胞由来のもので0.55cm及び0.12cmであり、iPS細胞由来のもので、1.37cm及び0.32cmであった。 As shown in FIG. 27, BRQ administration suppressed tumor growth. Although not shown in the figure, no visible side effect was observed in the BRQ-administered mice. As also shown in FIG. 28, teratomas size treated with DMSO and BRQ is each a 0.55 cm 3 and 0.12 cm 3 in those from ES cells, those derived from iPS cells, 1.37Cm 3 and It was 0.32 cm 3 .
 次に、それら腫瘍の切片を調製し、増殖マーカー Ki67、多能性幹細胞マーカー(Oct4及びNanog)、並びに分化マーカー(ATF,βIIIチューブリン及びSMA)を対象とする免疫標識を施し、観察した。 Next, sections of these tumors were prepared, subjected to immunolabeling with the proliferation marker Ki67, pluripotent stem cell markers (Oct4 and Nanog), and differentiation markers (ATF, βIII tubulin and SMA), and observed.
 その結果、図29に示すとおり、BRQ処理腫瘍におけるKi67陽性増殖細胞は、DMSO処理腫瘍におけるそれらと比較して、有意に減少した。また、図30に示すとおり、BRQ処理腫瘍において、Oct4陽性細胞及びNanog陽性細胞の数は、有意に減少したが、DMSO処理腫瘍において、多能性幹細胞マーカーを発現している細胞は多く認められた。一方、三胚葉における各マーカーであるATF,βIIIチューブリン及びSMAを各々発現する細胞を、両腫瘍とも同様に含んでいた。 As a result, as shown in FIG. 29, Ki67-positive proliferating cells in BRQ-treated tumors were significantly reduced as compared with those in DMSO-treated tumors. In addition, as shown in FIG. 30, the number of Oct4-positive cells and Nanog-positive cells was significantly reduced in BRQ-treated tumors, but many cells expressing pluripotent stem cell markers were observed in DMSO-treated tumors. It was On the other hand, both tumors similarly contained cells expressing ATF, βIII tubulin and SMA, which are markers in the three germ layers, respectively.
 したがって、DHODH阻害剤は、未分化の多能性幹細胞を特異的に除去することによって、分化細胞及びマウスに対して明白な細胞毒性をもたらすことなく、腫瘍形成を抑制できることが明らかになった。 Therefore, it was revealed that the DHODH inhibitor can suppress tumor formation by specifically removing undifferentiated pluripotent stem cells without causing obvious cytotoxicity to differentiated cells and mice.
 以上説明したように、本発明によれば、分化細胞には有意な細胞傷害をもたらすことなく、未分化の多能性幹細胞を除去することが可能となる。したがって、本発明は、多能性幹細胞から分化誘導した細胞群に残存する未分化の多能性幹細胞を除去することに優れているため、腫瘍化リスクが低減され、また副作用の少ない再生医療等において有用である。 As explained above, according to the present invention, it is possible to remove undifferentiated pluripotent stem cells without causing significant cytotoxicity to the differentiated cells. Therefore, the present invention is excellent in removing undifferentiated pluripotent stem cells remaining in a cell group differentiated from pluripotent stem cells, so that the risk of tumorigenesis is reduced, and regenerative medicine with few side effects, etc. Is useful in.

Claims (8)

  1.  ジヒドロオロト酸デヒドロゲナーゼ阻害剤を有効成分として含有する、多能性幹細胞から分化誘導した細胞群に残存する未分化の多能性幹細胞を除去するための組成物。 A composition for removing undifferentiated pluripotent stem cells remaining in a cell group differentiated from pluripotent stem cells, which contains a dihydroorotic acid dehydrogenase inhibitor as an active ingredient.
  2.  前記多能性幹細胞が、胚性幹細胞(ES細胞)、人工多能性幹細胞(iPS細胞)及び胚性腫瘍細胞(EC細胞)からなる群から選択される少なくとも1の多能性幹細胞である、請求項1に記載の組成物。 The pluripotent stem cells are at least one pluripotent stem cell selected from the group consisting of embryonic stem cells (ES cells), induced pluripotent stem cells (iPS cells) and embryonal tumor cells (EC cells), The composition according to claim 1.
  3.  前記ジヒドロオロト酸デヒドロゲナーゼ阻害剤がブレキナー(brequinar)である、請求項1又は2に記載の組成物。 The composition according to claim 1 or 2, wherein the dihydroorotic acid dehydrogenase inhibitor is brequinar.
  4.  前記細胞群が、前記多能性幹細胞から分化誘導した体性幹細胞を含む細胞群である、請求項1~3のうちのいずれか一項に記載の組成物。 The composition according to any one of claims 1 to 3, wherein the cell group is a cell group containing somatic stem cells differentiated from the pluripotent stem cells.
  5.  多能性幹細胞から分化誘導した細胞群とジヒドロオロト酸デヒドロゲナーゼ阻害剤とを接触させる工程を含む、前記細胞群から残存する未分化の多能性幹細胞を除去する方法。 A method for removing the remaining undifferentiated pluripotent stem cells from the cell group, which comprises the step of contacting a cell group differentiated from the pluripotent stem cell with a dihydroorotate dehydrogenase inhibitor.
  6.  前記多能性幹細胞が、胚性幹細胞(ES細胞)、人工多能性幹細胞(iPS細胞)及び胚性腫瘍細胞(EC細胞)からなる群から選択される少なくとも1の多能性幹細胞である、請求項5に記載の方法。 The pluripotent stem cells are at least one pluripotent stem cell selected from the group consisting of embryonic stem cells (ES cells), induced pluripotent stem cells (iPS cells) and embryonal tumor cells (EC cells), The method according to claim 5.
  7.  前記ジヒドロオロト酸デヒドロゲナーゼ阻害剤がブレキナー(brequinar)である、請求項5又は6に記載の方法。 The method according to claim 5 or 6, wherein the dihydroorotic acid dehydrogenase inhibitor is brequinar.
  8.  前記細胞群が、前記多能性幹細胞から分化誘導した体性幹細胞を含む細胞群である、請求項5~7のうちのいずれか一項に記載の方法。 The method according to any one of claims 5 to 7, wherein the cell group is a cell group containing somatic stem cells that are induced to differentiate from the pluripotent stem cells.
PCT/JP2019/049837 2018-12-21 2019-12-19 Composition for removing pluripotent stem cells and method of removing pluripotent stem cells WO2020130077A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US17/414,511 US20220364065A1 (en) 2018-12-21 2019-12-19 Composition for removing pluripotent stem cells and method of removing pluripotent stem cells
EP19898801.6A EP3901252A4 (en) 2018-12-21 2019-12-19 Composition for removing pluripotent stem cells and method of removing pluripotent stem cells
JP2020561509A JP7409670B2 (en) 2018-12-21 2019-12-19 Composition for removing pluripotent stem cells and method for removing pluripotent stem cells

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2018239318 2018-12-21
JP2018-239318 2018-12-21

Publications (1)

Publication Number Publication Date
WO2020130077A1 true WO2020130077A1 (en) 2020-06-25

Family

ID=71102823

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2019/049837 WO2020130077A1 (en) 2018-12-21 2019-12-19 Composition for removing pluripotent stem cells and method of removing pluripotent stem cells

Country Status (4)

Country Link
US (1) US20220364065A1 (en)
EP (1) EP3901252A4 (en)
JP (1) JP7409670B2 (en)
WO (1) WO2020130077A1 (en)

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008077639A1 (en) 2006-12-22 2008-07-03 Laboratorios Almirall, S.A. Amino nicotinic and isonicotinic acid derivatives as dhodh inhibitors
WO2012057052A1 (en) * 2010-10-25 2012-05-03 公立大学法人横浜市立大学 Use of peptidyl-prolyl isomerase pin1 for controlling stable maintenance and renewal of stem cells
JP2014501518A (en) * 2010-12-08 2014-01-23 ヴィアサイト,インコーポレイテッド Agents and methods for inhibiting the growth of human pluripotent stem cells
US20150306080A1 (en) * 2011-02-08 2015-10-29 Children's Medical Center Corporation Methods for treatment of melanoma
JP2017507649A (en) * 2014-01-21 2017-03-23 ザ メディカル カレッジ オブ ウィスコンシン インクThe Medical College Of Wisconsin, Inc. Methods for selective inhibition of pluripotent stem cells
JP2018014972A (en) * 2016-07-29 2018-02-01 国立大学法人大阪大学 Method for producing a differentiation-induced cell population from which undifferentiated cells are removed
WO2018110654A1 (en) * 2016-12-15 2018-06-21 Heartseed株式会社 Undifferentiated stem cell removal agent and method for removing undifferentiated stem cells

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3344243B1 (en) * 2015-09-01 2020-12-09 The Broad Institute, Inc. Compounds and methods useful for treating or preventing hematological cancers

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008077639A1 (en) 2006-12-22 2008-07-03 Laboratorios Almirall, S.A. Amino nicotinic and isonicotinic acid derivatives as dhodh inhibitors
WO2012057052A1 (en) * 2010-10-25 2012-05-03 公立大学法人横浜市立大学 Use of peptidyl-prolyl isomerase pin1 for controlling stable maintenance and renewal of stem cells
JP2014501518A (en) * 2010-12-08 2014-01-23 ヴィアサイト,インコーポレイテッド Agents and methods for inhibiting the growth of human pluripotent stem cells
US20150306080A1 (en) * 2011-02-08 2015-10-29 Children's Medical Center Corporation Methods for treatment of melanoma
JP2017507649A (en) * 2014-01-21 2017-03-23 ザ メディカル カレッジ オブ ウィスコンシン インクThe Medical College Of Wisconsin, Inc. Methods for selective inhibition of pluripotent stem cells
JP2018014972A (en) * 2016-07-29 2018-02-01 国立大学法人大阪大学 Method for producing a differentiation-induced cell population from which undifferentiated cells are removed
WO2018110654A1 (en) * 2016-12-15 2018-06-21 Heartseed株式会社 Undifferentiated stem cell removal agent and method for removing undifferentiated stem cells

Non-Patent Citations (25)

* Cited by examiner, † Cited by third party
Title
BEN-DAVID ET AL., CELL STEM CELL, vol. 12, 2013, pages 167 - 179
BIOCHEM J., vol. 336, no. 2, 1 December 1998 (1998-12-01), pages 299 - 303
BIOCHEMISTRY, vol. 47, no. 34, 26 August 2008 (2008-08-26), pages 8929 - 36
CAS, no. 717824-30-1
DEVELOPMENT, vol. 135, no. 17, September 2008 (2008-09-01), pages 2969 - 79
DIABETES, vol. 59, no. 9, September 2010 (2010-09-01), pages 2094 - 2101
EUR J MED CHEM., vol. 49, March 2012 (2012-03-01), pages 102 - 9
J MED CHEM., vol. 56, no. 8, 25 April 2013 (2013-04-25), pages 3148 - 67
JOHE KK ET AL., GENES DEV., vol. 10, no. 24, 1996, pages 3129 - 3140
KONDO ET AL., GENES DEV., vol. 18, no. 23, 2004, pages 2963 - 72
KONDO T.RAFF M., EMBO J., vol. 19, no. 9, 2000, pages 1998 - 2007
KUANG ET AL., CELL CHEM BIOL, vol. 24, 2017, pages 685 - 694
KURODA Y. ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 107, no. 19, 2010, pages 8639 - 8643
LADDS ET AL., NAT COMMUN, vol. 9, no. 1, 16 March 2018 (2018-03-16), pages 1107
NAT BIOTECHNOL., vol. 27, no. 3, March 2009 (2009-03-01), pages 275 - 80
NATURE, vol. 470, no. 7332, 3 February 2011 (2011-02-03), pages 105 - 9
NEURO ONCOL., 10 September 2019 (2019-09-10), pages nozl70
NISHIDE K ET AL., PLOS ONE, vol. 4, no. 8, 2009, pages e6869
PARR ET AL., SCI REP, vol. 6, 9 September 2016 (2016-09-09), pages 32532
See also references of EP3901252A4
SHIRAKI ET AL., CELL METAB, vol. 19, 2014, pages 780 - 794
SYKES ET AL., CELL, vol. 167, 2016, pages 171 - 186
TAKANAGA H ET AL., STEM CELLS, vol. 27, no. 1, 2009, pages 165 - 174
TANG ET AL., NAT BIOTECH, vol. 29, 2011, pages 829 - 834
TSUKAMOTO Y ET AL., STEM CELLS, vol. 34, no. 8, 2016, pages 2016 - 2025

Also Published As

Publication number Publication date
JPWO2020130077A1 (en) 2021-11-04
JP7409670B2 (en) 2024-01-09
EP3901252A4 (en) 2022-10-26
US20220364065A1 (en) 2022-11-17
EP3901252A1 (en) 2021-10-27

Similar Documents

Publication Publication Date Title
US20180273906A1 (en) Microvesicle and stem cell compositions for therapeutic applications
JP2022141694A (en) Compounds, compositions, methods and kits relating to telomere extension
US9410945B2 (en) Brown adipocyte progenitors in human skeletal muscle
Hatzistergos et al. S‐nitrosoglutathione reductase deficiency enhances the proliferative expansion of adult heart progenitors and myocytes post myocardial infarction
Ren et al. PKM2 regulates angiogenesis of VR‐EPCs through modulating glycolysis, mitochondrial fission, and fusion
Cárcamo-Orive et al. ERK2 protein regulates the proliferation of human mesenchymal stem cells without affecting their mobilization and differentiation potential
JP2010520181A (en) Osteopontin for the prediction and treatment of cardiovascular disease
US11873494B2 (en) Genetic and pharmacological transcriptional upregulation of the repressed FXN gene as a therapeutic strategy for Friedreich ataxia
Park et al. Vascular progenitors generated from tankyrase inhibitor-regulated naïve diabetic human iPSC potentiate efficient revascularization of ischemic retina
WO2018207952A1 (en) Composition for hair growth and/or hair restoration
Liu et al. Inhibition of mircoRNA-34a enhances survival of human bone marrow mesenchymal stromal/stem cells under oxidative stress
Begonja et al. Differential roles of cAMP and cGMP in megakaryocyte maturation and platelet biogenesis
Bekhite et al. Involvement of phosphoinositide 3-kinase class IA (PI3K 110α) and NADPH oxidase 1 (NOX1) in regulation of vascular differentiation induced by vascular endothelial growth factor (VEGF) in mouse embryonic stem cells
US20210275595A1 (en) Naïve human embryonic vascular progenitor cells and methods of treatment
JP7409670B2 (en) Composition for removing pluripotent stem cells and method for removing pluripotent stem cells
CA3154084A1 (en) Feeder-based and feeder-free stem cell culture systems for stratified epithelial stem cells, and uses related thereto
Kondo Selective eradication of pluripotent stem cells by inhibiting DHODH activity
Choi et al. Regulation of ROS-independent ERK signaling rescues replicative cellular senescence in ex vivo expanded human c-kit-positive cardiac progenitor cells
Hernández-García et al. Cell death activation during cavitation of embryoid bodies is mediated by hydrogen peroxide
KR101723292B1 (en) Composition comprising KAI1 polypeptide or nucleic acids encoding the same for inhibiting angiogenesis, and uses thereof
KR20180132298A (en) Use related with liver cell differentiation and liver disease using Yap/Taz
Matsushita et al. A novel oligodendrocyte cell line OLP6 shows the successive stages of oligodendrocyte development: late progenitor, immature and mature stages
Fabre et al. GREM1 is epigenetically reprogrammed in muscle cells after exercise training and controls myogenesis and metabolism
US20200299727A1 (en) Methods of treating danon disease
Huang et al. Brain isoform glycogen phosphorylase as a novel hepatic progenitor cell marker

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 19898801

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2020561509

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2019898801

Country of ref document: EP

Effective date: 20210721